Skip to main content
Erschienen in: Translational Neurodegeneration 1/2019

Open Access 01.12.2019 | Review

Beyond the synucleinopathies: alpha synuclein as a driving force in neurodegenerative comorbidities

verfasst von: Naomi P. Visanji, Anthony E. Lang, Gabor G. Kovacs

Erschienen in: Translational Neurodegeneration | Ausgabe 1/2019

Abstract

The fundamental role that alpha-synuclein (aSyn) plays in the pathogenesis of neurodegenerative synucleinopathies, including Parkinson’s disease, dementia with Lewy bodies, and multiple system atrophy, is a well-accepted fact. A wealth of experimental evidence has linked this relatively small but ubiquitously expressed protein to a plethora of cytopathologic mechanisms and suggests that aSyn may be capable of seeding the progressive spread of synucleinopathy throughout the brain. Beyond the synucleinopathies, the abnormal deposition of aSyn is frequently seen in a variety of other neurodegenerative proteinopathies including Alzheimer’s disease. In spite of the fact that the frequency of concomitant aSyn pathology in these disorders is such that it can be considered the rule rather than the exception, the potential role that aSyn may have in these disorders has received relatively little attention.
In this article we postulate that aSyn may in fact be a key protein in driving the pathogenic processes in neurodegenerative comorbidities. In addition to reviewing the frequency of concomitant deposition of aSyn in the neurodegenerative proteinopathies, we also consider our current understanding of the interaction of aSyn with other neurodegenerative disease-associated proteins, including tau, TDP-43, amyloid-β and prion protein, in the context of neuropathologic studies describing the anatomical sites of potential concomitant pathology. We conclude that a growing body of evidence, encompassing neuropathology studies in human brain, animal models of concomitant proteinopathies and studies employing sophisticated methods of probing protein-protein interaction, cumulatively suggest that aSyn is well positioned to exert a strong influence on the pathogenesis of the neurodegenerative comorbidities.
We hope to stimulate research in this emerging field and consider that future studies exploring the contribution of aSyn to the pathogenic processes in neurodegenerative comorbidities may provide critical information pertaining to diagnosis and the development of vital disease modifying treatments for these devastating diseases.
Abkürzungen
ARTAG
ageing-related tau astrogliopathy
aSyn
alpha-synuclein
amyloid-β
CGIs
glial cytoplasmic inclusions
DLB
dementia with Lewy bodies
FRET
Fluorescence Resonance Energy Transfer
FTLD
frontotemporal lobar degeneration
MSA
multiple system atrophy
PAF
pure autonomic failure
PART
primary age-related tauopathy
PD
Parkinson’s disease
PDD
Parkinson’s disease with dementia
PFFs
preformed fibrils
PrPC
cellular prior protein
TDP-43
transactive response DNA binding protein 43 kDa

Background

Alpha synuclein (aSyn) is a 14 kDa protein ubiquitously expressed in the presynaptic terminals of the brain, where it has been estimated to account for up to 1% of all cytosolic proteins [1, 2]. Since the discovery in 1997 that a mutation in SNCA, the gene that encodes aSyn, is linked to an autosomal dominant early-onset form of Parkinson’s disease (PD) [3] there has been an explosion of studies demonstrating the involvement of aSyn as a critical element of PD pathogenesis (Fig. 1). This more than of 20 years of research has yielded a wealth of evidence demonstrating that aggregated aSyn is a key feature of the neuropathology of PD and is heavily implicated in the neurodegenerative process in PD (reviewed in [4]). Some forms of aSyn aggregates can be neurotoxic and have been linked with a variety of deleterious effects in neurons including downregulation of mitochondrial complex1 activity [5], endoplasmic reticulum stress [6], neuroinflammation [7], disrupted cell membrane integrity [8] as well as inhibition of the ubiquitin proteasome system and impairment of the autophagy lysosomal pathway, which may in turn result in decreased degradation of aberrant aSyn, fueling a vicious neurotoxic cycle [9]. In addition, much recent attention has been paid to the observation that aSyn fibrils are seeds capable of inducing further aggregation of physiologic aSyn in a wide variety of model systems [10]. Collectively these findings have positioned aSyn at the epicenter PD research over the last 20 years. However, we are becoming increasingly aware that aSyn may also play a role in the pathogenesis of several other neurodegenerative proteinopathies.
There are several diseases in which pathologic aggregates of aSyn are a defining feature, collectively termed synucleinopathies. Primary synucleinopathies include Lewy body disorders, such as dementia with Lewy bodies (DLB), Parkinson’s disease (PD) PD with dementia (PDD) and pure autonomic failure (PAF). These are characterized by the predominance of intraneuronal cytoplasmic and neuritic deposits (Lewy bodies and Lewy neurites). The classification of these disorders is based on the clinical presentation and spatiotemporal development of aberrant aSyn pathology [11]. A further disorder, multiple system atrophy (MSA) is dominated by glial cytoplasmic inclusions (Papp-Lantos bodies). In addition to these primary synucleinopathies, deposition of aSyn is also commonly observed in the brains of individuals with other primary diagnoses (reviewed in [12, 13]). Aberrant accumulation of aSyn is frequently observed in brains with abnormal deposition of Tau, transactive response DNA binding protein 43 kDa (TDP-43), amyloid-β (Aβ) or prion protein. The frequency of these pathologic comorbidities is so common that their presence is the rule rather than the exception in neurodegenerative diseases. However, this frequency of co-occurrence is not reflected in published research, which has largely ignored this phenomenon. As illustrated in Fig. 1, although there are a large number of articles citing both aSyn and PD in the title, the number of articles citing aSyn in combination with either AD or tau in the title is disproportionately low.
Currently the pathological classification of neurodegenerative diseases is based on the predominant proteinopathy [11]. However, ignoring the existence of these comorbid proteinopathies that frequently exist simultaneously in the same brain likely impedes our understanding of disease pathogenesis, precludes the accurate early diagnosis of neurodegenerative proteinopathies and stratification of patients for clinical trials and jeopardizes the development of much needed disease modifying therapies. Here we review both the frequency of aSyn deposition in different neurodegenerative conditions and the available experimental studies on interaction of aSyn with other proteins associated with neurodegenerative disease with a focus on evidence suggesting aSyn may be a key protein in driving the neurodegenerative processes in these conditions. For clarity, it is our opinion that referring to the concomitant deposition of aSyn in diseases not classified as synucleinopathies as a “co-pathology” may infer a lesser importance of aSyn compared to the other abnormally deposited protein. We believe that, as the relative contribution of each aberrantly deposited protein to the neurodegenerative process is presently unknown, the term “concomitant pathology” is more appropriate. Concomitant is defined as “naturally accompanying or associated” reflecting both the frequency of this observation whilst remaining unbiased as to the relative importance of each aberrantly deposited protein to the neurodegenerative process as a whole.

Main text

Mechanisms of aSyn aggregation and propagation

Prior to considering any role aSyn may have in driving neuropathologic comorbidities it is important to appreciate the mechanisms by which aSyn has been shown to propagate synucleinopathy alone. This topic has been the focus of an impressive amount of research over the last decade inspired initially by the observation that aSyn pathology is evident in embryonic dopamine neurons grafted into the brains of human PD patients, leading to the hypothesis that aSyn may be capable of propagating synucleinopathy in a prion-like fashion [14]. There is now a wealth of experimental evidence ranging from studies in cultured cells to animal models demonstrating that aSyn seeds are capable of being transmitted from neuron to neuron and incorporating the aSyn of the host neuron into misfolded aggregates, leading to neuronal dysfunction and ultimately cell death (recently reviewed in [15]. Several studies interrogating the ability of aSyn to seed pathology in recipient cells or tissues have exploited preformed fibrils (PFFs) of misfolded aSyn generated by sonicating β-sheet rich fibrils of recombinant aSyn [16]. PFFs are rapidly taken up by numerous cell lines in culture, including primary dopamine neurons, whereby they induce the formation of aSyn aggregates bearing several similarities to Lewy bodies, including a high degree of phosphorylation at serine 129, polyubiquitination and coexpression of p62 [1619]. Similarly, inoculation of both wildtype and transgenic animals with PFFs results in the development of widespread synucleinopathy throughout synaptically connected networks, neurodegeneration and behavioural deficits (reviewed in [15, 20]. Other studies have demonstrated similar effects utilizing aSyn derived from human brain tissue from individuals with synucleinopathies [17, 21]. Identifying the mechanisms for uptake (putatively receptor mediated), processing (predominantly lysosomal) and release of these toxic aSyn seeds is the topic of ongoing research efforts (reviewed in [15]), as well as the possible contribution of different conformational strains of aSyn to the different pathological inclusions observed among the synucleinopathies. Thus, aSyn extracted from glial cytoplasmic inclusions has been shown to have a different proteolytic profile and a much more potent biological activity than that of Lewy body derived aSyn [21]. There are obvious therapeutic implications of this aspect of aSyn biology with several efforts underway to prevent aSyn spreading synucleinopathy throughout the brain, including the recent initiation of clinical trials of antibodies directed at aSyn. Certainly a better understanding of the mechanisms of release, uptake and trafficking of internalized aSyn should help provide several novel targets with disease modifying potential in PD and related synucleinopathies in the near future.

aSyn and comorbidity: a critical overview of neuropathological aspects

The association of synucleinopathy with different neurodegenerative conditions can be discussed from two perspectives. First, when aSyn appears as comorbidity (i.e., concurrent presence of brain disease with overlapping pathogenic aspects [13]) in diverse neurodegenerative conditions, and second when other proteinopathy comorbidities are observed in primary synucleinopathies. This is a somewhat arbitrary grouping as in cases with complex constellations of proteinopathies, it can be difficult to elucidate which is the predominant feature. In spite of the fact that emerging research is focusing on the description of concomitant pathologies, comparison between different studies will likely be hampered by the current lack of consensus regarding harmonization of nomenclature and evaluation strategies. There are several layers of complexity that must be kept in mind when evaluating the role of aSyn in proteinopathic comorbidities, thus simply referring to the presence of Lewy body pathology as a concomitant pathology might be insufficient for the reasons described below [13].
First, the classification of disorders with Lewy bodies awaits further crystallization. Diseases with Lewy bodies are grouped based on the early clinical presentation (movement disorder vs cognitive decline) [11]. However, although cluster analysis suggests that there are several distinct subtypes of PD [22], and several studies raise the concept that DLB might be distinct from PD and PDD [2325], there are currently no known biochemical or morphological features of aSyn pathologies allowing a clear division of subtypes of Lewy body disorders. Importantly, the anatomical distribution of synucleinopathy and the concomitant presence of further protein deposits (e.g., Aβ and tau-positive neurofibrillary tangles) might be an important aspect for distinguishing clinically different Lewy body disorders [23, 25]. Finally, there are conditions, detectable only by neuropathologic study, when Lewy bodies accumulate solely in the amygdala or the olfactory bulb or lower brainstem without any clinical manifestation (i.e, incidental Lewy body disease), or only in peripheral organs [11, 26]. Currently, two neuropathological approaches are used to describe Lewy-body related pathology. Braak staging of Lewy body pathology, delineates the sequential involvement of brain regions starting in the medulla oblongata (stage 1), pons (stage 2), mesencephalon, in particular the substantia nigra (stage 3), limbic areas (stage 4), and finally reaching neocortical areas (stages 5 and 6) [27]. Intriguingly, not all cases strictly follow the sequential distribution described by Braak staging [28]. A second set of criteria, which originates from the classification of Kosaka [29], are implemented in the diagnosis of DLB as either brainstem, limbic or neocortical types [30]. Since these approaches do not recognize the early or pure involvement of the olfactory bulb and various predominances of aSyn deposition, this was included in further strategy, the so called unified staging system for Lewy body disorders. This suggests classification of cases into one of the following stages: I. Olfactory Bulb Only; IIa Brainstem Predominant; IIb Limbic Predominant; III Brainstem and Limbic; IV Neocortical [31].
Second, historical studies focused exclusively on the presence of Lewy bodies and their clinicopathological relevance. However, the spectrum of aSyn accumulations in Lewy body disorders is much broader than the mere presence of Lewy bodies and involves depositions in synapses, neurites, astrocytes, and oligodendrocytes [11, 3234]. In addition, the use of modern techniques, such as the proximity ligation assay, has revealed further pathological alterations in neurons in Lewy body disorders [35]. Unfortunately, documentation of most of these aspects is lacking in the majority of existing clinicopathological studies, many of which also employed different antibodies for immunostaining, altogether jeopardizing our understanding of the role of aSyn pathology in diverse neurodegenerative conditions. These limitations call for a harmonized approach involving the evaluation of several anatomical regions using novel antibodies with standardized immunohistochemical, and other methods and a consensus description of anatomical patterns of different cellular or synaptic aSyn deposits to enable the comparison of different cohorts to enhance our understanding of the full spectrum of aSyn pathology in synucleinopathies.
The clinical subtypes associated with MSA pathology [36] cannot be clearly translated into biochemical or morphological differences. For the diagnosis of MSA the presence of oligodendroglial inclusions/glial cytoplasmic inclusions (CGIs) (Papp-Lantos bodies) is sufficient [37], however, neuronal aSyn pathology and further pathologies, described recently by sophisticated methods [38], should be also considered. The distribution of glial inclusions might follow either a striatonigral or olivopontocerebellar predominance or even be associated with frontotemporal lobar degeneration (FTLD) and prominent neuronal aSyn accumulation in the medial temporal lobe [39, 40]. Although incidental MSA cases are described [41], MSA is usually not sought in the diagnostic screening for concomitant pathologies since early steps of the disease might involve only the cerebellum [42]. Indeed, an aging study showed that by immunostaining several anatomical regions asymptomatic MSA cases could be identified in elderly communities [43].
Despite the relative frequency of concomitant neurodegenerative disorders, pure forms of proteinopathies are still apparent using current diagnostic methods. This supports the current protein-based molecular classification [13]. The interpretation of different studies is complicated by the fact that abnormally deposited proteins exist in different phases of aggregation or fibrillization, and in different phases or stages of sequential involvement of anatomical brain areas, which in turn are influenced by genetic variations, age and sex effects and multimorbidities including systemic and vascular disorders [13]. Furthermore, TDP-43 pathology has only recently been added to the spectrum of concomitant proteinopathies and only a very few studies report its frequency. In addition, tau pathology is perceived mostly as neurofibrillary tangles related to AD, thus, subcortical and astroglial tau pathology or even primary age-related tauopathy (PART) or the limbic tauopathy argyrophilic grain disease (AGD) is considerably underappreciated in most studies. These caveats have led to a wide range of reported frequencies of neurodegenerative comorbidities depending on diverse case collection and neuropathological methodological strategies, which we have summarised below.
TDP-43 pathology is clearly more frequent in Lewy body disorders (generally around 20%) [44], while less frequent in MSA (4–7%) [44, 45]. Further studies indicate that TDP-43 deposition in DLB (33.3%) is less frequent than in mixed AD/DLB cases (52.6%), or in AD (73.9%); but more frequent then in aged controls (17.9%) [46]. A recent study reported more extensive Lewy body distribution correlating with more frequent Aβ deposition in brainstem (50%), limbic (57%) and neocortical forms (80%) of Lewy body disorders or TDP-43 deposition (0, 16 and 22%, respectively) [44]. When comparing the clinical phenotypes, PDD and DLB are associated with more frequent AD-related pathology and TDP-43 proteinopathy than PD, but a similar frequency of AGD, and lower prevalence of PART [47]. These changes are associated with different loads of Aβ and tau pathologies in diverse anatomical regions [25]. Importantly, in MSA TDP-43 immunoreactivities comprise subpial astrocytic inclusions and glial cytoplasmic inclusions in addition to neuronal inclusions, dystrophic neurites, and perivascular inclusions [45]. Furthermore, concomitant Lewy bodies have been described in up to 10% of MSA cases [48]. Regarding AD-related pathology, intermediate to high levels of AD neuropathological changes have been described in ~ 8%, and PART in approximately 40% of MSA cases [44]. Any type of ageing-related tau astrogliopathy (ARTAG) is detected in up to 56% of synucleinopathy cases, specifically grey matter ARTAG has been reported in 36.8% of Lewy body disorders and 17.1% of MSA cases [49]. In a small group of neuropathological controls (i.e. lack of any neurodegenerative disease entity including PART) any type of ARTAG was observed in 28.6% and grey matter ARTAG was lacking (0%) [49].
On the other hand, Lewy body pathology is reported in a wide range of neurodegenerative disorders and in unimpaired aging (Fig. 2). Studies in ageing cohorts, irrespective whether the individuals showed neurological symptoms or not, have typically focused on the presence of Lewy bodies related to AD-related pathological change and have reported frequencies ranging from 6 to 39% [12]. Studies employing various methods have suggested a frequency of concomitant Lewy bodies in ~ 20% in CBD and PSP, which is thought to be only slightly higher than that in the general aged population [44, 50, 51]. A somewhat lower prevalence of concomitant Lewy body pathology has been reported in FTLD-TDP and ALS-TDP cases (11–15%) [44] and in sporadic Creutzfeldt-Jakob disease (prion disease) (9–23%) [52, 53]. In comparison the prevalence of Lewy pathology in unimpaired aging has been reported as ~ 13% in a single study [54].

Experimental studies on the interaction of aSyn with other neurodegenerative disease-associated proteins

1)
Tau
 
Experimental studies suggest that aSyn and tau, including oligomeric forms, may directly interact with each other, furthermore, co-deposition of both proteins in Lewy bodies, Lewy neurites and tangles has been shown using antibodies against different epitopes of tau and aSyn in postmortem tissues (reviewed in [55]). The latter depends on the anatomical location and disease type exemplified by the relative paucity of co-localization in the substantia nigra in concomitant progressive supranuclear palsy and Lewy body pathology, whereas it is more likely to detect co-localization in the amygdala in concomitant AD and Lewy body pathology [5658]. Whereas aSyn has a tendency to self-aggregate [59], the aggregation of tau relies on a variety of cofactors, many of which likely remain to be identified [60]. A seminal study by Giasson and colleagues implicated aSyn as a possible cofactor in the aggregation of tau [61]. Using recombinant aSyn and tau these authors showed a synergistic relationship whereby one protein can act to cross-seed the aggregation of the other in vitro. More recent studies in cultured cells, whilst supporting the ability of recombinant aSyn fibrils to induce aggregation of tau, however, have cast some doubt on the reciprocal ability of tau fibrils to cross-seed the aggregation of aSyn [62]. These discrepancies may reflect the disparate techniques used to determine interaction between these two proteins. Indeed, one study using Fluorescence Resonance Energy Transfer (FRET) microscopy, failed to show any cross-seeding between aSyn and tau [63]. This method together with proximity ligation assay also failed to provide clear evidence on the close proximity allowing direct interaction of tau and aSyn in human brain tissue in spite of rare co-localization in inclusion bodies in the amygdala [56]. Alternatively, these discrepancies may relate to variability in the capability of different conformational strains, or different antibodies raised against distinct biochemical modifications of these proteins in human studies, a further source of variability between studies, to interact with each other, as previously demonstrated by Guo and colleagues [64].
The observation that individuals from the Contursi kindred, who express the aggregation prone A53T mutation in aSyn, also have tau filamentous amyloid inclusions, as do mice that overexpress A53T or E46K mutant aSyn, provides evidence that mutant aSyn can drive aggregation of tau in vivo [61, 65, 66]. Furthermore, affinity chromatography in human brain lysates has revealed a direct interaction between the C terminus of wildtype aSyn and the microtubule binding domain of tau [67]. In addition, mice injected with recombinant preformed fibrils (PFFs) of wildtype aSyn develop not only a widespread synucleinopathy but also develop tau deposits, further supporting a synergistic relationship between aSyn and tau aggregation in vivo [68].
Interestingly, in an experimental setting there is little evidence of overlap between tau and aSyn deposits in either cultured cells or in mice exposed to aSyn PFFs [62, 69]. Furthermore, as tauopathy is frequently found in the absence of synucleinopathy in human brain [70], although evidence suggests aSyn may well be a co-factor able to drive the aggregation of tau, many further studies should be undertaken to determine the magnitude of any such effect in human comorbid proteinopathies.
2)
TDP-43
 
TDP-43 is frequently associated with tau and Aβ, yet to date only two recent studies, from the same group, have addressed the interaction between aSyn and TDP-43 at an experimental level. SHSY5Y cells, double transfected with both aSyn and a mutant TDP-43 lacking the nuclear localization signal (TDP-43dNLS) developed abundant aggregates of phosphorylated TDP-43 upon exposure to aSyn PFFs [62]. These aggregates were not formed in cells expressing wildtype TDP-43, suggesting that the predominantly cytosolic TDP-43dNLS is more susceptible to the aggregation catalysing effects of aSyn PFFs than the predominantly nuclear wildtype TDP-43. In a second in vivo study, wildtype mice inoculated with intracerebral aSyn PFFs developed a synucleinopathy as well as abnormal dot-like TDP-43 deposits [68]. Similar to the distribution of PFF-induced synucleinopathy and tau deposition, inclusions of aSyn and TDP-43 were only partly colocalized in cells exposed to aSyn PFFs and rarely colocalized in mice exposed to aSyn PFFs calling into question the significance of any synergistic effect of aSyn on TDP-43 aggregation in disease [62, 68]. In human tissue rare co-localizations are observed for TDP-43 in amygdala Lewy bodies or aSyn in amygdala TDP-43 inclusions [56], while in MSA–related oligodendroglial aSyn inclusions occasionally TDP-43 immunoreactivity can be observed [45].
3)
Amyloid-β
 
In 1993 Ueda and colleagues reported a study on an unrecognized component of amyloid in AD and tentatively named this 35-amino acid peptide NAC (non-Aβ component of AD amyloid) and its precursor NACP [71], which soon was defined as aSyn [1, 2]. Later it was shown that the amyloid core of Aβ plaques itself lacks aSyn deposition [72] but the protein is seen in the dystrophic neurites of plaques. These observations in human tissue sparked interest in examining these two proteins as potential interactors.
Indeed, experimental studies provide a wealth of evidence thought to support the binding and coaggregation of Aβ and aSyn. In vitro, Aβ can co-seed the aggregation of aSyn in both cell free and cell-based systems [7378] and recombinant Aβ can induce phosphorylation of aSyn at ser 129 (aSyn-ser129P) [79]. In animal models, there is conflicting evidence regarding the interaction between aSyn and Aβ. Thus, the combined expression of human Aβ and aSyn has been shown to increase the deposition of intraneuronal fibrillar aSyn and accelerate the development of both motor and cognitive dysfunction in double transgenic mice [73]. Similarly in a mouse model of DLB-AD, that simultaneously expresses PS1(M146 V), APP(Swe), tau(P301L) and aSyn(A53T), a substantial increase in deposition of Aβ, tau and aSyn was observed, accompanied by accelerated cognitive decline, suggestive of a synergistic effect of these proteins in driving both pathology and phenotype [80]. Supporting this possible synergistic effect, aSyn knock-down in an APP transgenic mouse model of AD has been shown to reduce the degeneration of cholinergic fibers and hippocampal neurons [81]. Interestingly these effects were in the absence of any effect on either APP expression or Aβ deposition, and may point to an as yet unknown influence of aSyn on the selective vulnerability of cholinergic neurons in AD. Together these studies provide evidence supporting the provocative hypothesis that aSyn may be more than merely an idle passenger in AD pathogenesis. In contrast, some studies have suggested that aSyn may inhibit the formation of Aβ plaques, with one demonstrating a significant increase in Aβ plaque load in APP (Tg2576) mice when crossed with aSyn knockout mice [82], whilst a second study found that aSyn knockdown in APP mice increased plaque burden but decreased levels of extracellular Aβ oligomers [83]. A further study found that inoculation of APPPS1/aSyn(A30P) mice with brain homogenate derived aSyn reduced the formation of Aβ plaques and that the seeding capacity of Aβ brain homogenates was significantly reduced in the presence of aSyn [84].
Recent studies have also suggested a further, albeit indirect mechanism by which aSyn may mediate deposition of Aβ. Following the observation that exposure to recombinant aSyn leads to increased levels of Aβ in cultured PC12 cells or primary hippocampal neurons [85, 86] a recent report has suggested that aSyn may induce the production and secretion of Aβ through enhanced cleavage of APP in cultured neuronal cells [87]. It is tempting to speculate that this observation could underly the frequently observed accumulation of Aβ in synucleinopathies if so, then probing the precise mechanisms by which aSyn mediate the processing of APP could prove invaluable in the development of potentially disease-modifying therapeutics.
4)
Prion protein
 
The cellular prior protein (PrPC) has previously been suggested as a receptor for Aβ oligomers [88, 89] although others have reported evidence to the contrary [90]. Similarly, cell surface PrPC has also been described as a putative receptor that promotes the uptake of aSyn via binding to the N-terminal domain [91, 92], although others have more recently disputed these claims [89]. Thus, Aulic and colleagues reported that PrPC knockdown in murine neuroblastoma cells attenuated the uptake of recombinant aSyn oligomers with a similar effect noted when comparing aSyn uptake in mouse primary hippocampal neurons prepared from wildtype or PrPC knockout mice. In addition, PrPC knockout mice developed lower levels of aSyn aggregates in the cortex, striatum, thalamus and hippocampus following intrastriatal inoculation with aSyn fibrils, suggesting that PrPC may facilitate the uptake and aggregation of aSyn oligomers. These same authors provided evidence that the replication of scrapie prions was blocked by aSyn oligomers [93] providing a possible explanation for the observation that individuals with Creutzfeldt-Jakob disease have a more protracted disease course when there is concomitant synucleinopathy [94]. Conversely, La Vitola and colleagues found no evidence of binding between PrPC and aSyn oligomers [89] and noted that both PrPC knockout mice and wildtype mice were equally susceptible to aSyn oligomer-mediated toxicity and that PrPC expression was not a prerequisite for these toxic effects. It remains possible that these discrepancies are a result of methodological differences between the studies. Certainly, there are many different species of aSyn oligomers, which may have a differential binding capacity with PrPC and it remains possible that future studies could reveal that both PrPC -dependent and PrPC -independent pathways play a role in synucleinopathies [95]. On the other hand, aSyn has an unexpected role in inducing a transmissible spongiform encephalopathy with accumulation of disease-associated PrP [96]. Accordingly, aggregated aSyn is potent in cross-seeding of prion protein misfolding and aggregation in vitro, producing self-replicating states that can lead to transmissible prion diseases upon serial passaging in wild type animals.

Anatomical sites of potential concomitant pathology in the human brain

An understanding of the typical anatomical deposition profiles of different neurodegenerative disease-associated proteins throughout the brain is vital to considering the possible impact of aSyn on the pathogenesis of other comorbid proteinopathies. As an example, Fig. 3 illustrates the predicted overlap of aSyn in PD (the most common synucleinopathy) with deposition patterns for Aβ in AD (the most common neurodegenerative proteinopathy) and Fig. 4 illustrates the predicted overlap of aSyn in PD with tau in AD/PART [97101]. In the majority of cases, according to Braak et al. [97], in the early stages (1 to 3) of PD aSyn is predominantly deposited in the brainstem, then progresses through the limbic (including amygdala) and subcortical (including basal ganglia) regions, eventually reaching the neocortical areas in later stages of the disease. In AD, however, the pattern of Aβ deposition is essentially the opposite, with deposits first observed in the neocortex (i.e. Thal phase 1) then in the limbic and subcortical regions (i.e. Thal Phase 2 and 3) and finally being found also in the brainstem (Thal Phase 4) progressing to the cerebellum in Thal Phase 5 [102]. Thus, the opportunity for aSyn and Aβ to coexist in the same anatomical region of the brain does not present until at least one of the comorbidities is at an advanced stage. The region of the brain in which co-deposition may occur is dependent on the relative stage of each disease. Thus, an individual with early PD, but more advanced AD-related pathology would be predicted to have overlapping pathology in the brainstem only (Fig. 3). Conversely an individual with early AD, but a more advanced PD would have overlapping pathology only in the neocortex (Fig. 3). Individuals with advanced AD and PD would have the maximum potential for concomitant pathologies throughout the brain.
With respect to co-occurence of tau in AD/PART and aSyn in PD, the opportunity for the two proteins to be deposited in the same anatomical region occurs first in the brainstem, in particular the locus coeruelus, in early stages of disease, indicated as subcortical stages of neurofibrillary tangle pathology by Braak and colleagues [100] (Fig. 4). The deposition of proteins then progresses to limbic and subcortical regions. Since tau pathology (Braak stages I-IV) predominates in the limbic areas and the medial temporal lobe but not in the basal ganglia while Lewy body pathology (Braak stage 4) in the basal ganglia and limbic areas, the most likely meeting point would be the amygdala, hippocampus, entorhinal cortex and and anterior cingulate. Finally, at more advances stages the two proteins may also coexist in the neocortex (Fig. 4).
Irrespective of which proteins are co-deposited, clearly, the degree of potential overlap is dependent on the temporal stage of each disease, with fewer overlapping regions predicted early in disease and a much higher degree of potential overlap in later stages, thus emphasizing the potential contribution of comorbid proteinopathies to pathogenicity particularly in later disease. It is also important to note that these figures highlight the potential brain regions where one would predict comorbid proteinopathies to coexist, but remain agnostic to the amount of pathology present, or which if any proteinopathy is predominant. Finally, we emphasize that these images (Figs. 3 and 4) represent our knowledge based on current immunohistochemical methods to detect abnormal pathology. Hence, it remains possible that future studies mapping aberrantly deposited proteins using more sensitive and sophisticated methods, may reveal a yet unexpected and greater degree of anatomical overlap.
On an anatomical level the amygdala, a part of the limbic system located deep in the medial temporal lobe, suffers a high degree of overlapping aberrantly deposited proteins with pathology present in AD, synucleinopathies and tauopathies among others [58]. This observation has led Nelson and colleagues to recently propose that the amygdala may serve as an “incubator” for misfolded proteins and suggest that the convergence of neuropathologic comorbidities in this regions may act in synergy to drive the evolution of a pure proteinopathy to potentially more aggressive neuropathologic comorbidities [58]. This provocative hypothesis, if true, would have profound implications for both the diagnosis of neurodegenerative diseases as well as the development of novel therapeutics targeting proteinopathy. Over the next few years we hope that, using modern techniques able to probe the biochemical properties of aberrant proteins, the careful study of the common neuropathologic comorbidities, particularly in brain regions with a high degree of convergence, will provide vital information regarding the relationship between these comorbidities, the impact each may impart on the other and potentially reveal novel targets for future biomarkers and disease modifying therapies.

Conclusions

Given the frequency of occurrence, the impact of comorbid disease-associated proteins on disease pathogenesis in neurodegenerative proteinopathies has to date been relatively underexplored. It remains possible that the aggregation of multiple disease associated proteins in a given condition are unrelated to each other, perhaps the result of a common deleterious process in the brain, for example neuroinflammation or disturbances of the protein processing systems. However, emerging evidence suggests that the concomitant deposition of multiple disease-associated proteins may impact upon disease pathogenesis and have clinical significance. Based on neuropathological studies, as depicted in Fig. 5, the presence of comorbid proteinopathies, aSyn (depicted in green), AD-related pathology (depicted in yellow) and TDP-43 (depicted in orange), increase in tandem with each other. Thus, in premotor PD, Braak stages 1 & 2, there is minimal comorbid AD-related pathology or deposition of TDP-43. As PD progresses to a motor stage the concomitant deposition of both AD-related pathology and TDP-43 to increase in parallel, with the highest levels of comorbidity observed in PDD and DLB which encompass cognitive symptoms. Indeed, the existence of comorbid conditions often leads to a significant overlap in symptoms. For example, the clinical spectrum of parkinsonism is associated with the involvement of anatomical regions beyond those important for the organization of movement which in turn are affected by a wide variety of other neuropathologically defined disorders and pathologically altered proteins. Therefore, the clinical symptoms alone do not predict incontrovertibly the underlying molecular pathology. This circumstance jeopardizes the accurate early diagnosis of neurodegenerative proteinopathies and stratification of patients for clinical trials. Thus, even though the methods and interpretations of these aforementioned studies on comorbid proteinopathies vary considerably, on the whole these support the rationale for developing highly specific markers of disease-associated proteins involved in neurodegenerative conditions and particularly for examining all of these together in neurodegenerative conditions. Defining clusters of patients based on the patterns of comorbid proteinopathies will enhance research in disease pathogenesis, lead to an improved prognostic predictive value, and therefore, may be useful for stratifying patients for clinical trial and monitoring efficacy of novel therapies [103].
In this review we speculate that aSyn may be an as yet unrecognised potentiator of neurodegenerative diseases beyond those formally recognized as synucleinopathies. aSyn is bestowed with several characteristics that may allow this protein to impact the pathogenesis of multiple neurodegenerative proteinopathies. First, aSyn is abundantly expressed in the synapses of the brain and indeed, it is named after its localization on synaptic vesicles. All neurodegenerative diseases involve synapses (reviewed in [104]). Thus, it is very tempting to speculate that aSyn may be implicated in the synaptic dysfunction that is a feature shared by all neurodegenerative proteinopathies irrespective of the predominant pathology. Second, although disease-associated aSyn deposits are intracellular (Lewy bodies, Lewy neurites, glial cytoplasmic inclusions), aSyn and its neurotoxic oligomers are highly soluble, can be excreted into the extracellular space and are widely distributed throughout the brain [10]. This feature, (shared with tau, TDP-43 and prion protein), allows aSyn unrestricted access to both intracellular and extracellular compartments of the brain and thus permits aSyn to impart widespread effects. Third, although other neurodegenerative disease-associated proteins have been shown capable of seeing proteinopathy, it has been recently shown that aSyn may be particularly potent at cross-seeding the aggregation of other disease-associated proteins [96].
In addition to Lewy bodies or abnormally aggregated aSyn, detectable with current immunohistochemical methods, aSyn may also undergo more subtle biochemical alterations, such as phosphorylation. These changes can occur in human brains with or without neurodegenerative proteinopathies and may also precede the development of Lewy bodies [54, 105, 106]. Thus, soluble aSyn levels have been found to be approximately doubled in AD in the absence of synucleinopathy, and to more closely correlate with the degree of cognitive impairment than soluble Aβ or tau [107]. Furthermore, it is well established that aSyn can have detrimental effect on synaptic biology, in the absence of synucleinopathy [108110]. These observations suggest that by focusing only on aggregated aSyn we may underestimate role of aSyn in comorbidities and it could perhaps be a silent potentiator driving disease. Further studies investigating the molecular and biochemical features of synaptosomal aSyn in AD, for example, have the potential to reveal that aSyn may have a more widespread role in neurodegenerative diseases beyond the synucleinopathies. If born out, this would have clear implications for the development of disease modifying therapies for neurodegenerative diseases. For example, aSyn immunotherapy could have an attenuating effect on AD-related pathology and symptoms for example. Such a hypothesis is certainly supported by the observation that co-expression of aSyn and Aβ induces a more aggressive cognitive decline in AD [80].
It is important to note, however, that the evidence linking aSyn to other neurodegenerative proteinopathies is presently largely correlative. Indeed, it remains possible that, being an aggregation prone protein, biochemical and cellular changes in other neurodegenerative proteinopathies trigger coincidental aggregation of aSyn. Thus, until such studies have been undertaken as to provide direct evidence supporting that aSyn is the driving force for other neurodegenerative proteinopathies caution should be exercised in interpreting clinicopathologic observations.
In conclusion, it is likely that future disease modifying therapies for neurodegenerative proteinopathies will entail a precision-based approach tailored to the molecular underpinning of disease in each patient. In order to achieve this goal we first need a better understanding of the interaction of different proteins in comorbid proteinopathies. This information will be vital to support effective stratification of patients for clinical trials and eventually for the successful application of disease appropriate therapeutic interventions.

Acknowledgements

The authors would like to acknowledge the Edmond J. Safra Program in Parkinson’s Disease and Rossy Program for PSP research.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Jakes R, Spillantini MG, Goedert M. Identification of two distinct synucleins from human brain. FEBS Lett. 1994;345(1):27–32.PubMedCrossRef Jakes R, Spillantini MG, Goedert M. Identification of two distinct synucleins from human brain. FEBS Lett. 1994;345(1):27–32.PubMedCrossRef
2.
Zurück zum Zitat Iwai A, Masliah E, Yoshimoto M, Ge N, Flanagan L, de Silva HA, et al. The precursor protein of non-a beta component of Alzheimer's disease amyloid is a presynaptic protein of the central nervous system. Neuron. 1995;14(2):467–75.PubMedCrossRef Iwai A, Masliah E, Yoshimoto M, Ge N, Flanagan L, de Silva HA, et al. The precursor protein of non-a beta component of Alzheimer's disease amyloid is a presynaptic protein of the central nervous system. Neuron. 1995;14(2):467–75.PubMedCrossRef
3.
Zurück zum Zitat Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science. 1997;276(5321):2045–7.PubMedCrossRef Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science. 1997;276(5321):2045–7.PubMedCrossRef
5.
Zurück zum Zitat Devi L, Raghavendran V, Prabhu BM, Avadhani NG, Anandatheerthavarada HK. Mitochondrial import and accumulation of alpha-synuclein impair complex I in human dopaminergic neuronal cultures and Parkinson disease brain. J Biol Chem. 2008;283(14):9089–100.PubMedPubMedCentralCrossRef Devi L, Raghavendran V, Prabhu BM, Avadhani NG, Anandatheerthavarada HK. Mitochondrial import and accumulation of alpha-synuclein impair complex I in human dopaminergic neuronal cultures and Parkinson disease brain. J Biol Chem. 2008;283(14):9089–100.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Colla E, Jensen PH, Pletnikova O, Troncoso JC, Glabe C, Lee MK. Accumulation of toxic alpha-synuclein oligomer within endoplasmic reticulum occurs in synucleinopathy in vivo. J Neurosci. 2012;32(10):3301–5.PubMedPubMedCentralCrossRef Colla E, Jensen PH, Pletnikova O, Troncoso JC, Glabe C, Lee MK. Accumulation of toxic alpha-synuclein oligomer within endoplasmic reticulum occurs in synucleinopathy in vivo. J Neurosci. 2012;32(10):3301–5.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Zhang W, Wang T, Pei Z, Miller DS, Wu X, Block ML, et al. Aggregated alpha-synuclein activates microglia: a process leading to disease progression in Parkinson's disease. FASEB J. 2005;19(6):533–42.PubMedCrossRef Zhang W, Wang T, Pei Z, Miller DS, Wu X, Block ML, et al. Aggregated alpha-synuclein activates microglia: a process leading to disease progression in Parkinson's disease. FASEB J. 2005;19(6):533–42.PubMedCrossRef
8.
Zurück zum Zitat Danzer KM, Haasen D, Karow AR, Moussaud S, Habeck M, Giese A, et al. Different species of alpha-synuclein oligomers induce calcium influx and seeding. J Neurosci. 2007;27(34):9220–32.PubMedPubMedCentralCrossRef Danzer KM, Haasen D, Karow AR, Moussaud S, Habeck M, Giese A, et al. Different species of alpha-synuclein oligomers induce calcium influx and seeding. J Neurosci. 2007;27(34):9220–32.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Xilouri M, Brekk OR, Stefanis L. Alpha-Synuclein and protein degradation systems: a reciprocal relationship. Mol Neurobiol. 2013;47(2):537–51.PubMedCrossRef Xilouri M, Brekk OR, Stefanis L. Alpha-Synuclein and protein degradation systems: a reciprocal relationship. Mol Neurobiol. 2013;47(2):537–51.PubMedCrossRef
10.
Zurück zum Zitat Karpowicz RJ Jr, Trojanowski JQ, Lee VM. Transmission of alpha-synuclein seeds in neurodegenerative disease: recent developments. Lab Investig. 2019;99(7):971–981.PubMedCrossRef Karpowicz RJ Jr, Trojanowski JQ, Lee VM. Transmission of alpha-synuclein seeds in neurodegenerative disease: recent developments. Lab Investig. 2019;99(7):971–981.PubMedCrossRef
11.
Zurück zum Zitat Kovacs GG. Molecular Pathological Classification of Neurodegenerative Diseases: Turning towards Precision Medicine. Int J Mol Sci. 2016;17(2):E189.PubMedCentralCrossRef Kovacs GG. Molecular Pathological Classification of Neurodegenerative Diseases: Turning towards Precision Medicine. Int J Mol Sci. 2016;17(2):E189.PubMedCentralCrossRef
13.
Zurück zum Zitat Kovacs GG. Are comorbidities compatible with a molecular pathological classification of neurodegenerative diseases? Curr Opin Neurol. 2019;32(2):279–91.PubMedCrossRef Kovacs GG. Are comorbidities compatible with a molecular pathological classification of neurodegenerative diseases? Curr Opin Neurol. 2019;32(2):279–91.PubMedCrossRef
14.
Zurück zum Zitat Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, et al. Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host-to-graft disease propagation. Nat Med. 2008;14(5):501–3.PubMedCrossRef Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, et al. Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host-to-graft disease propagation. Nat Med. 2008;14(5):501–3.PubMedCrossRef
15.
Zurück zum Zitat Henderson MX, Trojanowski JQ, Lee VM. Alpha-Synuclein pathology in Parkinson's disease and related synucleinopathies. Neurosci Lett. 2019;709:134316.PubMedCrossRefPubMedCentral Henderson MX, Trojanowski JQ, Lee VM. Alpha-Synuclein pathology in Parkinson's disease and related synucleinopathies. Neurosci Lett. 2019;709:134316.PubMedCrossRefPubMedCentral
16.
Zurück zum Zitat Volpicelli-Daley LA, Luk KC, Patel TP, Tanik SA, Riddle DM, Stieber A, et al. Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron. 2011;72:57–71.PubMedPubMedCentralCrossRef Volpicelli-Daley LA, Luk KC, Patel TP, Tanik SA, Riddle DM, Stieber A, et al. Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron. 2011;72:57–71.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Henderson MX, Peng C, Trojanowski JQ, Lee VMY. LRRK2 activity does not dramatically alter alpha-synuclein pathology in primary neurons. Acta Neuropathol Commun. 2018;6(1):45.PubMedPubMedCentralCrossRef Henderson MX, Peng C, Trojanowski JQ, Lee VMY. LRRK2 activity does not dramatically alter alpha-synuclein pathology in primary neurons. Acta Neuropathol Commun. 2018;6(1):45.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Henderson MX, Chung CH, Riddle DM, Zhang B, Gathagan RJ, Seeholzer SH, et al. Unbiased proteomics of early Lewy body formation model implicates active microtubule affinity-regulating kinases (MARKs) in Synucleinopathies. J Neurosci. 2017;37(24):5870–84.PubMedPubMedCentralCrossRef Henderson MX, Chung CH, Riddle DM, Zhang B, Gathagan RJ, Seeholzer SH, et al. Unbiased proteomics of early Lewy body formation model implicates active microtubule affinity-regulating kinases (MARKs) in Synucleinopathies. J Neurosci. 2017;37(24):5870–84.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Dryanovski DI, Guzman JN, Xie Z, Galteri DJ, Volpicelli-Daley LA, Lee VM, et al. Calcium entry and alpha-synuclein inclusions elevate dendritic mitochondrial oxidant stress in dopaminergic neurons. J Neurosci. 2013;33(24):10154–64.PubMedPubMedCentralCrossRef Dryanovski DI, Guzman JN, Xie Z, Galteri DJ, Volpicelli-Daley LA, Lee VM, et al. Calcium entry and alpha-synuclein inclusions elevate dendritic mitochondrial oxidant stress in dopaminergic neurons. J Neurosci. 2013;33(24):10154–64.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Visanji NP, Brotchie JM, Kalia LV, Koprich JB, Tandon A, Watts JC, et al. Alpha-Synuclein-based animal models of Parkinson's disease: challenges and opportunities in a new era. Trends Neurosci. 2016;39(11):750–62.PubMedCrossRef Visanji NP, Brotchie JM, Kalia LV, Koprich JB, Tandon A, Watts JC, et al. Alpha-Synuclein-based animal models of Parkinson's disease: challenges and opportunities in a new era. Trends Neurosci. 2016;39(11):750–62.PubMedCrossRef
21.
Zurück zum Zitat Peng C, Gathagan RJ, Covell DJ, Medellin C, Stieber A, Robinson JL, et al. Cellular milieu imparts distinct pathological alpha-synuclein strains in synucleinopathies. Nature. 2018;557(7706):558–63.PubMedPubMedCentralCrossRef Peng C, Gathagan RJ, Covell DJ, Medellin C, Stieber A, Robinson JL, et al. Cellular milieu imparts distinct pathological alpha-synuclein strains in synucleinopathies. Nature. 2018;557(7706):558–63.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat van Rooden SM, Heiser WJ, Kok JN, Verbaan D, van Hilten JJ, Marinus J. The identification of Parkinson's disease subtypes using cluster analysis: a systematic review. Mov Disord. 2010;25(8):969–78.PubMedCrossRef van Rooden SM, Heiser WJ, Kok JN, Verbaan D, van Hilten JJ, Marinus J. The identification of Parkinson's disease subtypes using cluster analysis: a systematic review. Mov Disord. 2010;25(8):969–78.PubMedCrossRef
23.
Zurück zum Zitat Halliday GM, Holton JL, Revesz T, Dickson DW. Neuropathology underlying clinical variability in patients with synucleinopathies. Acta Neuropathol. 2011;122(2):187–204.PubMedCrossRef Halliday GM, Holton JL, Revesz T, Dickson DW. Neuropathology underlying clinical variability in patients with synucleinopathies. Acta Neuropathol. 2011;122(2):187–204.PubMedCrossRef
24.
Zurück zum Zitat Irwin DJ, Lee VM, Trojanowski JQ. Parkinson's disease dementia: convergence of alpha-synuclein, tau and amyloid-beta pathologies. Nat Rev Neurosci. 2013;14(9):626–36.PubMedPubMedCentralCrossRef Irwin DJ, Lee VM, Trojanowski JQ. Parkinson's disease dementia: convergence of alpha-synuclein, tau and amyloid-beta pathologies. Nat Rev Neurosci. 2013;14(9):626–36.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Jellinger KA. Dementia with Lewy bodies and Parkinson's disease-dementia: current concepts and controversies. J Neural Transm (Vienna). 2018;125(4):615–50.CrossRef Jellinger KA. Dementia with Lewy bodies and Parkinson's disease-dementia: current concepts and controversies. J Neural Transm (Vienna). 2018;125(4):615–50.CrossRef
26.
Zurück zum Zitat Orimo S, Ghebremedhin E, Gelpi E. Peripheral and central autonomic nervous system: does the sympathetic or parasympathetic nervous system bear the brunt of the pathology during the course of sporadic PD? Cell Tissue Res. 2018;373(1):267–86.PubMedCrossRef Orimo S, Ghebremedhin E, Gelpi E. Peripheral and central autonomic nervous system: does the sympathetic or parasympathetic nervous system bear the brunt of the pathology during the course of sporadic PD? Cell Tissue Res. 2018;373(1):267–86.PubMedCrossRef
27.
Zurück zum Zitat Gispert S, Del Turco D, Garrett L, Chen A, Bernard DJ, Hamm-Clement J, et al. Transgenic mice expressing mutant A53T human alpha-synuclein show neuronal dysfunction in the absence of aggregate formation. Mol Cell Neurosci. 2003;24(2):419–29.PubMedCrossRef Gispert S, Del Turco D, Garrett L, Chen A, Bernard DJ, Hamm-Clement J, et al. Transgenic mice expressing mutant A53T human alpha-synuclein show neuronal dysfunction in the absence of aggregate formation. Mol Cell Neurosci. 2003;24(2):419–29.PubMedCrossRef
28.
Zurück zum Zitat Jellinger KA. A critical reappraisal of current staging of Lewy-related pathology in human brain. Acta Neuropathol. 2008;116(1):1–16.PubMedCrossRef Jellinger KA. A critical reappraisal of current staging of Lewy-related pathology in human brain. Acta Neuropathol. 2008;116(1):1–16.PubMedCrossRef
29.
Zurück zum Zitat Kosaka K, Tsuchiya K, Yoshimura M. Lewy body disease with and without dementia: a clinicopathological study of 35 cases. Clin Neuropathol. 1988;7(6):299–305.PubMed Kosaka K, Tsuchiya K, Yoshimura M. Lewy body disease with and without dementia: a clinicopathological study of 35 cases. Clin Neuropathol. 1988;7(6):299–305.PubMed
30.
Zurück zum Zitat McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor JP, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies: fourth consensus report of the DLB consortium. Neurology. 2017;89(1):88–100.PubMedPubMedCentralCrossRef McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor JP, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies: fourth consensus report of the DLB consortium. Neurology. 2017;89(1):88–100.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Beach TG, Adler CH, Lue L, Sue LI, Bachalakuri J, Henry-Watson J, et al. Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta Neuropathol. 2009;117(6):613–34.PubMedPubMedCentralCrossRef Beach TG, Adler CH, Lue L, Sue LI, Bachalakuri J, Henry-Watson J, et al. Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta Neuropathol. 2009;117(6):613–34.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Rohan Z, Milenkovic I, Lutz MI, Matej R, Kovacs GG. Shared and distinct patterns of Oligodendroglial response in synucleinopathies and Tauopathies. J Neuropathol Exp Neurol. 2016;75(12):1100–9.PubMedCrossRef Rohan Z, Milenkovic I, Lutz MI, Matej R, Kovacs GG. Shared and distinct patterns of Oligodendroglial response in synucleinopathies and Tauopathies. J Neuropathol Exp Neurol. 2016;75(12):1100–9.PubMedCrossRef
33.
Zurück zum Zitat Colom-Cadena M, Pegueroles J, Herrmann AG, Henstridge CM, Munoz L, Querol-Vilaseca M, et al. Synaptic phosphorylated alpha-synuclein in dementia with Lewy bodies. Brain. 2017;140(12):3204–14.PubMedPubMedCentralCrossRef Colom-Cadena M, Pegueroles J, Herrmann AG, Henstridge CM, Munoz L, Querol-Vilaseca M, et al. Synaptic phosphorylated alpha-synuclein in dementia with Lewy bodies. Brain. 2017;140(12):3204–14.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Schulz-Schaeffer WJ. The synaptic pathology of alpha-synuclein aggregation in dementia with Lewy bodies, Parkinson's disease and Parkinson's disease dementia. Acta Neuropathol. 2010;120(2):131–43.PubMedPubMedCentralCrossRef Schulz-Schaeffer WJ. The synaptic pathology of alpha-synuclein aggregation in dementia with Lewy bodies, Parkinson's disease and Parkinson's disease dementia. Acta Neuropathol. 2010;120(2):131–43.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Roberts RF, Wade-Martins R, Alegre-Abarrategui J. Direct visualization of alpha-synuclein oligomers reveals previously undetected pathology in Parkinson's disease brain. Brain. 2015;138(Pt 6):1642–57.PubMedPubMedCentralCrossRef Roberts RF, Wade-Martins R, Alegre-Abarrategui J. Direct visualization of alpha-synuclein oligomers reveals previously undetected pathology in Parkinson's disease brain. Brain. 2015;138(Pt 6):1642–57.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Jellinger KA. Neuropathology of multiple system atrophy: new thoughts about pathogenesis. Mov Disord. 2014;29(14):1720–41.PubMedCrossRef Jellinger KA. Neuropathology of multiple system atrophy: new thoughts about pathogenesis. Mov Disord. 2014;29(14):1720–41.PubMedCrossRef
37.
Zurück zum Zitat Trojanowski JQ, Revesz T. Neuropathology working group on MSA. Proposed neuropathological criteria for the post mortem diagnosis of multiple system atrophy. Neuropathol Appl Neurobiol. 2007;33(6):615–20.PubMedCrossRef Trojanowski JQ, Revesz T. Neuropathology working group on MSA. Proposed neuropathological criteria for the post mortem diagnosis of multiple system atrophy. Neuropathol Appl Neurobiol. 2007;33(6):615–20.PubMedCrossRef
38.
Zurück zum Zitat Sekiya H, Kowa H, Koga H, Takata M, Satake W, Futamura N, et al. Wide distribution of alpha-synuclein oligomers in multiple system atrophy brain detected by proximity ligation. Acta Neuropathol. 2019;137(3):455–66.PubMedCrossRef Sekiya H, Kowa H, Koga H, Takata M, Satake W, Futamura N, et al. Wide distribution of alpha-synuclein oligomers in multiple system atrophy brain detected by proximity ligation. Acta Neuropathol. 2019;137(3):455–66.PubMedCrossRef
39.
Zurück zum Zitat Aoki N, Boyer PJ, Lund C, Lin WL, Koga S, Ross OA, et al. Atypical multiple system atrophy is a new subtype of frontotemporal lobar degeneration: frontotemporal lobar degeneration associated with alpha-synuclein. Acta Neuropathol. 2015;130(1):93–105.PubMedCrossRefPubMedCentral Aoki N, Boyer PJ, Lund C, Lin WL, Koga S, Ross OA, et al. Atypical multiple system atrophy is a new subtype of frontotemporal lobar degeneration: frontotemporal lobar degeneration associated with alpha-synuclein. Acta Neuropathol. 2015;130(1):93–105.PubMedCrossRefPubMedCentral
40.
Zurück zum Zitat Rohan Z, Rahimi J, Weis S, Kapas I, Auff E, Mitrovic N, et al. Screening for alpha-synuclein immunoreactive neuronal inclusions in the hippocampus allows identification of atypical MSA (FTLD-synuclein). Acta Neuropathol. 2015;130(2):299–301.PubMedCrossRef Rohan Z, Rahimi J, Weis S, Kapas I, Auff E, Mitrovic N, et al. Screening for alpha-synuclein immunoreactive neuronal inclusions in the hippocampus allows identification of atypical MSA (FTLD-synuclein). Acta Neuropathol. 2015;130(2):299–301.PubMedCrossRef
41.
42.
Zurück zum Zitat Brettschneider J, Irwin DJ, Boluda S, Byrne MD, Fang L, Lee EB, et al. Progression of alpha-synuclein pathology in multiple system atrophy of the cerebellar type. Neuropathol Appl Neurobiol. 2017;43(4):315–29.PubMedCrossRef Brettschneider J, Irwin DJ, Boluda S, Byrne MD, Fang L, Lee EB, et al. Progression of alpha-synuclein pathology in multiple system atrophy of the cerebellar type. Neuropathol Appl Neurobiol. 2017;43(4):315–29.PubMedCrossRef
43.
Zurück zum Zitat Kovacs GG, Milenkovic I, Wohrer A, Hoftberger R, Gelpi E, Haberler C, et al. Non-Alzheimer neurodegenerative pathologies and their combinations are more frequent than commonly believed in the elderly brain: a community-based autopsy series. Acta Neuropathol. 2013;126(3):365–84.PubMedCrossRef Kovacs GG, Milenkovic I, Wohrer A, Hoftberger R, Gelpi E, Haberler C, et al. Non-Alzheimer neurodegenerative pathologies and their combinations are more frequent than commonly believed in the elderly brain: a community-based autopsy series. Acta Neuropathol. 2013;126(3):365–84.PubMedCrossRef
44.
Zurück zum Zitat Robinson JL, Lee EB, Xie SX, Rennert L, Suh E, Bredenberg C, et al. Neurodegenerative disease concomitant proteinopathies are prevalent, age-related and APOE4-associated. Brain. 2018;141(7):2181–93.PubMedPubMedCentralCrossRef Robinson JL, Lee EB, Xie SX, Rennert L, Suh E, Bredenberg C, et al. Neurodegenerative disease concomitant proteinopathies are prevalent, age-related and APOE4-associated. Brain. 2018;141(7):2181–93.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Koga S, Lin WL, Walton RL, Ross OA, Dickson DW. TDP-43 pathology in multiple system atrophy: colocalization of TDP-43 and alpha-synuclein in glial cytoplasmic inclusions. Neuropathol Appl Neurobiol. 2018;44(7):707–21.PubMedCrossRefPubMedCentral Koga S, Lin WL, Walton RL, Ross OA, Dickson DW. TDP-43 pathology in multiple system atrophy: colocalization of TDP-43 and alpha-synuclein in glial cytoplasmic inclusions. Neuropathol Appl Neurobiol. 2018;44(7):707–21.PubMedCrossRefPubMedCentral
46.
Zurück zum Zitat McAleese KE, Walker L, Erskine D, Thomas AJ, McKeith IG, Attems J. TDP-43 pathology in Alzheimer's disease, dementia with Lewy bodies and ageing. Brain Pathol. 2017;27(4):472–9.PubMedCrossRef McAleese KE, Walker L, Erskine D, Thomas AJ, McKeith IG, Attems J. TDP-43 pathology in Alzheimer's disease, dementia with Lewy bodies and ageing. Brain Pathol. 2017;27(4):472–9.PubMedCrossRef
47.
Zurück zum Zitat Colom-Cadena M, Grau-Rivera O, Planellas L, Cerquera C, Morenas E, Helgueta S, et al. Regional overlap of pathologies in Lewy body disorders. J Neuropathol Exp Neurol. 2017;76(3):216–24.PubMed Colom-Cadena M, Grau-Rivera O, Planellas L, Cerquera C, Morenas E, Helgueta S, et al. Regional overlap of pathologies in Lewy body disorders. J Neuropathol Exp Neurol. 2017;76(3):216–24.PubMed
48.
Zurück zum Zitat Koga S, Parks A, Uitti RJ, van Gerpen JA, Cheshire WP, Wszolek ZK, et al. Profile of cognitive impairment and underlying pathology in multiple system atrophy. Mov Disord. 2017;32(3):405–13.PubMedCrossRef Koga S, Parks A, Uitti RJ, van Gerpen JA, Cheshire WP, Wszolek ZK, et al. Profile of cognitive impairment and underlying pathology in multiple system atrophy. Mov Disord. 2017;32(3):405–13.PubMedCrossRef
49.
Zurück zum Zitat Kovacs GG, Robinson JL, Xie SX, Lee EB, Grossman M, Wolk DA, et al. Evaluating the patterns of aging-related tau Astrogliopathy unravels novel insights into brain aging and neurodegenerative diseases. J Neuropathol Exp Neurol. 2017;76(4):270–88.PubMedPubMedCentralCrossRef Kovacs GG, Robinson JL, Xie SX, Lee EB, Grossman M, Wolk DA, et al. Evaluating the patterns of aging-related tau Astrogliopathy unravels novel insights into brain aging and neurodegenerative diseases. J Neuropathol Exp Neurol. 2017;76(4):270–88.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Dugger BN, Adler CH, Shill HA, Caviness J, Jacobson S, Driver-Dunckley E, et al. Concomitant pathologies among a spectrum of parkinsonian disorders. Parkinsonism Relat Disord. 2014;20(5):525–9.PubMedPubMedCentralCrossRef Dugger BN, Adler CH, Shill HA, Caviness J, Jacobson S, Driver-Dunckley E, et al. Concomitant pathologies among a spectrum of parkinsonian disorders. Parkinsonism Relat Disord. 2014;20(5):525–9.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Tsuboi Y, Ahlskog JE, Apaydin H, Parisi JE, Dickson DW. Lewy bodies are not increased in progressive supranuclear palsy compared with normal controls. Neurology. 2001;57(9):1675–8.PubMedCrossRef Tsuboi Y, Ahlskog JE, Apaydin H, Parisi JE, Dickson DW. Lewy bodies are not increased in progressive supranuclear palsy compared with normal controls. Neurology. 2001;57(9):1675–8.PubMedCrossRef
52.
Zurück zum Zitat Grau-Rivera O, Gelpi E, Nos C, Gaig C, Ferrer I, Saiz A, et al. Clinicopathological correlations and concomitant pathologies in rapidly progressive dementia: a brain Bank series. Neurodegener Dis. 2015;15(6):350–60.PubMedCrossRef Grau-Rivera O, Gelpi E, Nos C, Gaig C, Ferrer I, Saiz A, et al. Clinicopathological correlations and concomitant pathologies in rapidly progressive dementia: a brain Bank series. Neurodegener Dis. 2015;15(6):350–60.PubMedCrossRef
53.
Zurück zum Zitat Vital A, Fernagut PO, Canron MH, Joux J, Bezard E, Martin-Negrier ML, et al. The nigrostriatal pathway in Creutzfeldt-Jakob disease. J Neuropathol Exp Neurol. 2009;68(7):809–15.PubMedCrossRef Vital A, Fernagut PO, Canron MH, Joux J, Bezard E, Martin-Negrier ML, et al. The nigrostriatal pathway in Creutzfeldt-Jakob disease. J Neuropathol Exp Neurol. 2009;68(7):809–15.PubMedCrossRef
54.
Zurück zum Zitat Parkkinen L, Soininen H, Alafuzoff I. Regional distribution of alpha-synuclein pathology in unimpaired aging and Alzheimer disease. J Neuropathol Exp Neurol. 2003;62(4):363–7.PubMedCrossRef Parkkinen L, Soininen H, Alafuzoff I. Regional distribution of alpha-synuclein pathology in unimpaired aging and Alzheimer disease. J Neuropathol Exp Neurol. 2003;62(4):363–7.PubMedCrossRef
55.
Zurück zum Zitat Moussaud S, Jones DR, Moussaud-Lamodiere EL, Delenclos M, Ross OA, McLean PJ. Alpha-synuclein and tau: teammates in neurodegeneration? Mol Neurodegener. 2014;9:43.PubMedPubMedCentralCrossRef Moussaud S, Jones DR, Moussaud-Lamodiere EL, Delenclos M, Ross OA, McLean PJ. Alpha-synuclein and tau: teammates in neurodegeneration? Mol Neurodegener. 2014;9:43.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Lutz MI, Schwaiger C, Hochreiter B, Kovacs GG, Schmid JA. Novel approach for accurate tissue-based protein colocalization and proximity microscopy. Sci Rep. 2017;7(1):2668.PubMedPubMedCentralCrossRef Lutz MI, Schwaiger C, Hochreiter B, Kovacs GG, Schmid JA. Novel approach for accurate tissue-based protein colocalization and proximity microscopy. Sci Rep. 2017;7(1):2668.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Uchikado H, DelleDonne A, Ahmed Z, Dickson DW. Lewy bodies in progressive supranuclear palsy represent an independent disease process. J Neuropathol Exp Neurol. 2006;65(4):387–95.PubMedCrossRef Uchikado H, DelleDonne A, Ahmed Z, Dickson DW. Lewy bodies in progressive supranuclear palsy represent an independent disease process. J Neuropathol Exp Neurol. 2006;65(4):387–95.PubMedCrossRef
58.
Zurück zum Zitat Nelson PT, Abner EL, Patel E, Anderson S, Wilcock DM, Kryscio RJ, et al. The amygdala as a locus of pathologic Misfolding in neurodegenerative diseases. J Neuropathol Exp Neurol. 2018;77(1):2–20.PubMedCrossRef Nelson PT, Abner EL, Patel E, Anderson S, Wilcock DM, Kryscio RJ, et al. The amygdala as a locus of pathologic Misfolding in neurodegenerative diseases. J Neuropathol Exp Neurol. 2018;77(1):2–20.PubMedCrossRef
59.
Zurück zum Zitat Giasson BI, Uryu K, Trojanowski JQ, Lee VM. Mutant and wild type human alpha-synucleins assemble into elongated filaments with distinct morphologies in vitro. J Biol Chem. 1999;274(12):7619–22.PubMedCrossRef Giasson BI, Uryu K, Trojanowski JQ, Lee VM. Mutant and wild type human alpha-synucleins assemble into elongated filaments with distinct morphologies in vitro. J Biol Chem. 1999;274(12):7619–22.PubMedCrossRef
60.
Zurück zum Zitat Goedert M, Jakes R, Spillantini MG, Hasegawa M, Smith MJ, Crowther RA. Assembly of microtubule-associated protein tau into Alzheimer-like filaments induced by sulphated glycosaminoglycans. Nature. 1996;383(6600):550–3.PubMedCrossRef Goedert M, Jakes R, Spillantini MG, Hasegawa M, Smith MJ, Crowther RA. Assembly of microtubule-associated protein tau into Alzheimer-like filaments induced by sulphated glycosaminoglycans. Nature. 1996;383(6600):550–3.PubMedCrossRef
61.
Zurück zum Zitat Giasson BI, Forman MS, Higuchi M, Golbe LI, Graves CL, Kotzbauer PT, et al. Initiation and synergistic fibrillization of tau and alpha-synuclein. Science. 2003;300(5619):636–40.PubMedCrossRef Giasson BI, Forman MS, Higuchi M, Golbe LI, Graves CL, Kotzbauer PT, et al. Initiation and synergistic fibrillization of tau and alpha-synuclein. Science. 2003;300(5619):636–40.PubMedCrossRef
62.
Zurück zum Zitat Nonaka T, Masuda-Suzukake M, Hasegawa M. Molecular mechanisms of the co-deposition of multiple pathological proteins in neurodegenerative diseases. Neuropathology. 2018;38(1):64–71.PubMedCrossRef Nonaka T, Masuda-Suzukake M, Hasegawa M. Molecular mechanisms of the co-deposition of multiple pathological proteins in neurodegenerative diseases. Neuropathology. 2018;38(1):64–71.PubMedCrossRef
63.
Zurück zum Zitat Holmes BB, Furman JL, Mahan TE, Yamasaki TR, Mirbaha H, Eades WC, et al. Proteopathic tau seeding predicts tauopathy in vivo. Proc Natl Acad Sci U S A. 2014;111(41):E4376–85.PubMedPubMedCentralCrossRef Holmes BB, Furman JL, Mahan TE, Yamasaki TR, Mirbaha H, Eades WC, et al. Proteopathic tau seeding predicts tauopathy in vivo. Proc Natl Acad Sci U S A. 2014;111(41):E4376–85.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Guo JL, Covell DJ, Daniels JP, Iba M, Stieber A, Zhang B, et al. Distinct alpha-synuclein strains differentially promote tau inclusions in neurons. Cell. 2013;154(1):103–17.PubMedCrossRef Guo JL, Covell DJ, Daniels JP, Iba M, Stieber A, Zhang B, et al. Distinct alpha-synuclein strains differentially promote tau inclusions in neurons. Cell. 2013;154(1):103–17.PubMedCrossRef
65.
Zurück zum Zitat Emmer KL, Waxman EA, Covy JP, Giasson BI. E46K human alpha-synuclein transgenic mice develop Lewy-like and tau pathology associated with age-dependent, detrimental motor impairment. J Biol Chem. 2011;286(40):35104–18.PubMedPubMedCentralCrossRef Emmer KL, Waxman EA, Covy JP, Giasson BI. E46K human alpha-synuclein transgenic mice develop Lewy-like and tau pathology associated with age-dependent, detrimental motor impairment. J Biol Chem. 2011;286(40):35104–18.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Duda JE, Giasson BI, Mabon ME, Miller DC, Golbe LI, Lee VM, et al. Concurrence of alpha-synuclein and tau brain pathology in the Contursi kindred. Acta Neuropathol. 2002;104(1):7–11.PubMedCrossRef Duda JE, Giasson BI, Mabon ME, Miller DC, Golbe LI, Lee VM, et al. Concurrence of alpha-synuclein and tau brain pathology in the Contursi kindred. Acta Neuropathol. 2002;104(1):7–11.PubMedCrossRef
67.
Zurück zum Zitat Jensen PH, Hager H, Nielsen MS, Hojrup P, Gliemann J, Jakes R. Alpha-synuclein binds to tau and stimulates the protein kinase A-catalyzed tau phosphorylation of serine residues 262 and 356. J Biol Chem. 1999;274(36):25481–9.PubMedCrossRef Jensen PH, Hager H, Nielsen MS, Hojrup P, Gliemann J, Jakes R. Alpha-synuclein binds to tau and stimulates the protein kinase A-catalyzed tau phosphorylation of serine residues 262 and 356. J Biol Chem. 1999;274(36):25481–9.PubMedCrossRef
68.
Zurück zum Zitat Masuda-Suzukake M, Nonaka T, Hosokawa M, Kubo M, Shimozawa A, Akiyama H, et al. Pathological alpha-synuclein propagates through neural networks. Acta Neuropathol Commun. 2014;2:88.PubMedPubMedCentralCrossRef Masuda-Suzukake M, Nonaka T, Hosokawa M, Kubo M, Shimozawa A, Akiyama H, et al. Pathological alpha-synuclein propagates through neural networks. Acta Neuropathol Commun. 2014;2:88.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Oikawa T, Nonaka T, Terada M, Tamaoka A, Hisanaga S, Hasegawa M. Alpha-Synuclein fibrils exhibit gain of toxic function, promoting tau aggregation and inhibiting microtubule assembly. J Biol Chem. 2016;291(29):15046–56.PubMedPubMedCentralCrossRef Oikawa T, Nonaka T, Terada M, Tamaoka A, Hisanaga S, Hasegawa M. Alpha-Synuclein fibrils exhibit gain of toxic function, promoting tau aggregation and inhibiting microtubule assembly. J Biol Chem. 2016;291(29):15046–56.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Thal DR, von Arnim CA, Griffin WS, Mrak RE, Walker L, Attems J, et al. Frontotemporal lobar degeneration FTLD-tau: preclinical lesions, vascular, and Alzheimer-related co-pathologies. J Neural Transm (Vienna). 2015;122(7):1007–18.CrossRef Thal DR, von Arnim CA, Griffin WS, Mrak RE, Walker L, Attems J, et al. Frontotemporal lobar degeneration FTLD-tau: preclinical lesions, vascular, and Alzheimer-related co-pathologies. J Neural Transm (Vienna). 2015;122(7):1007–18.CrossRef
71.
Zurück zum Zitat Ueda K, Fukushima H, Masliah E, Xia Y, Iwai A, Yoshimoto M, et al. Molecular cloning of cDNA encoding an unrecognized component of amyloid in Alzheimer disease. Proc Natl Acad Sci U S A. 1993;90(23):11282–6.PubMedPubMedCentralCrossRef Ueda K, Fukushima H, Masliah E, Xia Y, Iwai A, Yoshimoto M, et al. Molecular cloning of cDNA encoding an unrecognized component of amyloid in Alzheimer disease. Proc Natl Acad Sci U S A. 1993;90(23):11282–6.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Bayer TA, Jakala P, Hartmann T, Havas L, McLean C, Culvenor JG, et al. Alpha-synuclein accumulates in Lewy bodies in Parkinson's disease and dementia with Lewy bodies but not in Alzheimer's disease beta-amyloid plaque cores. Neurosci Lett. 1999;266(3):213–6.PubMedCrossRef Bayer TA, Jakala P, Hartmann T, Havas L, McLean C, Culvenor JG, et al. Alpha-synuclein accumulates in Lewy bodies in Parkinson's disease and dementia with Lewy bodies but not in Alzheimer's disease beta-amyloid plaque cores. Neurosci Lett. 1999;266(3):213–6.PubMedCrossRef
73.
Zurück zum Zitat Masliah E, Rockenstein E, Veinbergs I, Sagara Y, Mallory M, Hashimoto M, et al. Beta-amyloid peptides enhance alpha-synuclein accumulation and neuronal deficits in a transgenic mouse model linking Alzheimer's disease and Parkinson's disease. Proc Natl Acad Sci U S A. 2001;98(21):12245–50.PubMedPubMedCentralCrossRef Masliah E, Rockenstein E, Veinbergs I, Sagara Y, Mallory M, Hashimoto M, et al. Beta-amyloid peptides enhance alpha-synuclein accumulation and neuronal deficits in a transgenic mouse model linking Alzheimer's disease and Parkinson's disease. Proc Natl Acad Sci U S A. 2001;98(21):12245–50.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Ono K, Takahashi R, Ikeda T, Yamada M. Cross-seeding effects of amyloid beta-protein and alpha-synuclein. J Neurochem. 2012;122(5):883–90.PubMedCrossRef Ono K, Takahashi R, Ikeda T, Yamada M. Cross-seeding effects of amyloid beta-protein and alpha-synuclein. J Neurochem. 2012;122(5):883–90.PubMedCrossRef
75.
Zurück zum Zitat Mandal PK, Pettegrew JW, Masliah E, Hamilton RL, Mandal R. Interaction between Abeta peptide and alpha synuclein: molecular mechanisms in overlapping pathology of Alzheimer's and Parkinson's in dementia with Lewy body disease. Neurochem Res. 2006;31(9):1153–62.PubMedCrossRef Mandal PK, Pettegrew JW, Masliah E, Hamilton RL, Mandal R. Interaction between Abeta peptide and alpha synuclein: molecular mechanisms in overlapping pathology of Alzheimer's and Parkinson's in dementia with Lewy body disease. Neurochem Res. 2006;31(9):1153–62.PubMedCrossRef
76.
Zurück zum Zitat Atsmon-Raz Y, Miller Y. Non-amyloid-beta component of human alpha-Synuclein oligomers induces formation of new Abeta oligomers: insight into the mechanisms that link Parkinson's and Alzheimer's diseases. ACS Chem Neurosci. 2016;7(1):46–55.PubMedCrossRef Atsmon-Raz Y, Miller Y. Non-amyloid-beta component of human alpha-Synuclein oligomers induces formation of new Abeta oligomers: insight into the mechanisms that link Parkinson's and Alzheimer's diseases. ACS Chem Neurosci. 2016;7(1):46–55.PubMedCrossRef
77.
Zurück zum Zitat Tsigelny IF, Crews L, Desplats P, Shaked GM, Sharikov Y, Mizuno H, et al. Mechanisms of hybrid oligomer formation in the pathogenesis of combined Alzheimer's and Parkinson's diseases. PLoS One. 2008;3(9):e3135.PubMedPubMedCentralCrossRef Tsigelny IF, Crews L, Desplats P, Shaked GM, Sharikov Y, Mizuno H, et al. Mechanisms of hybrid oligomer formation in the pathogenesis of combined Alzheimer's and Parkinson's diseases. PLoS One. 2008;3(9):e3135.PubMedPubMedCentralCrossRef
78.
79.
Zurück zum Zitat Swirski M, Miners JS, de Silva R, Lashley T, Ling H, Holton J, et al. Evaluating the relationship between amyloid-beta and alpha-synuclein phosphorylated at Ser129 in dementia with Lewy bodies and Parkinson's disease. Alzheimers Res Ther. 2014;6(5–8):77.PubMedPubMedCentralCrossRef Swirski M, Miners JS, de Silva R, Lashley T, Ling H, Holton J, et al. Evaluating the relationship between amyloid-beta and alpha-synuclein phosphorylated at Ser129 in dementia with Lewy bodies and Parkinson's disease. Alzheimers Res Ther. 2014;6(5–8):77.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Clinton LK, Blurton-Jones M, Myczek K, Trojanowski JQ, LaFerla FM. Synergistic interactions between Abeta, tau, and alpha-synuclein: acceleration of neuropathology and cognitive decline. J Neurosci. 2010;30(21):7281–9.PubMedPubMedCentralCrossRef Clinton LK, Blurton-Jones M, Myczek K, Trojanowski JQ, LaFerla FM. Synergistic interactions between Abeta, tau, and alpha-synuclein: acceleration of neuropathology and cognitive decline. J Neurosci. 2010;30(21):7281–9.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Spencer B, Desplats PA, Overk CR, Valera-Martin E, Rissman RA, Wu C, et al. Reducing endogenous alpha-Synuclein mitigates the degeneration of selective neuronal populations in an Alzheimer's disease transgenic mouse model. J Neurosci. 2016;36(30):7971–84.PubMedPubMedCentralCrossRef Spencer B, Desplats PA, Overk CR, Valera-Martin E, Rissman RA, Wu C, et al. Reducing endogenous alpha-Synuclein mitigates the degeneration of selective neuronal populations in an Alzheimer's disease transgenic mouse model. J Neurosci. 2016;36(30):7971–84.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Kallhoff V, Peethumnongsin E, Zheng H. Lack of alpha-synuclein increases amyloid plaque accumulation in a transgenic mouse model of Alzheimer's disease. Mol Neurodegener. 2007;2:6.PubMedPubMedCentralCrossRef Kallhoff V, Peethumnongsin E, Zheng H. Lack of alpha-synuclein increases amyloid plaque accumulation in a transgenic mouse model of Alzheimer's disease. Mol Neurodegener. 2007;2:6.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Khan SS, LaCroix M, Boyle G, Sherman MA, Brown JL, Amar F, et al. Bidirectional modulation of Alzheimer phenotype by alpha-synuclein in mice and primary neurons. Acta Neuropathol. 2018;136(4):589–605.PubMedCrossRefPubMedCentral Khan SS, LaCroix M, Boyle G, Sherman MA, Brown JL, Amar F, et al. Bidirectional modulation of Alzheimer phenotype by alpha-synuclein in mice and primary neurons. Acta Neuropathol. 2018;136(4):589–605.PubMedCrossRefPubMedCentral
84.
Zurück zum Zitat Bachhuber T, Katzmarski N, McCarter JF, Loreth D, Tahirovic S, Kamp F, et al. Inhibition of amyloid-beta plaque formation by alpha-synuclein. Nat Med. 2015;21(7):802–7.PubMedCrossRef Bachhuber T, Katzmarski N, McCarter JF, Loreth D, Tahirovic S, Kamp F, et al. Inhibition of amyloid-beta plaque formation by alpha-synuclein. Nat Med. 2015;21(7):802–7.PubMedCrossRef
85.
Zurück zum Zitat Kazmierczak A, Strosznajder JB, Adamczyk A. Alpha-Synuclein enhances secretion and toxicity of amyloid beta peptides in PC12 cells. Neurochem Int. 2008;53(6–8):263–9.PubMedCrossRef Kazmierczak A, Strosznajder JB, Adamczyk A. Alpha-Synuclein enhances secretion and toxicity of amyloid beta peptides in PC12 cells. Neurochem Int. 2008;53(6–8):263–9.PubMedCrossRef
86.
Zurück zum Zitat Majd S, Chegini F, Chataway T, Zhou XF, Gai W. Reciprocal induction between alpha-synuclein and beta-amyloid in adult rat neurons. Neurotox Res. 2013;23(1):69–78.PubMedCrossRef Majd S, Chegini F, Chataway T, Zhou XF, Gai W. Reciprocal induction between alpha-synuclein and beta-amyloid in adult rat neurons. Neurotox Res. 2013;23(1):69–78.PubMedCrossRef
87.
Zurück zum Zitat Roberts HL, Schneider BL, Brown DR. alpha-Synuclein increases beta-amyloid secretion by promoting beta−/gamma-secretase processing of APP. PloS one. 2017;12(2):e0171925.PubMedPubMedCentralCrossRef Roberts HL, Schneider BL, Brown DR. alpha-Synuclein increases beta-amyloid secretion by promoting beta−/gamma-secretase processing of APP. PloS one. 2017;12(2):e0171925.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Lauren J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmatter SM. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature. 2009;457(7233):1128–32.PubMedPubMedCentralCrossRef Lauren J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmatter SM. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature. 2009;457(7233):1128–32.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat La Vitola P, Beeg M, Balducci C, Santamaria G, Restelli E, Colombo L, et al. Cellular prion protein neither binds to alpha-synuclein oligomers nor mediates their detrimental effects. Brain. 2019;142(2):249–54.PubMedCrossRef La Vitola P, Beeg M, Balducci C, Santamaria G, Restelli E, Colombo L, et al. Cellular prion protein neither binds to alpha-synuclein oligomers nor mediates their detrimental effects. Brain. 2019;142(2):249–54.PubMedCrossRef
90.
91.
Zurück zum Zitat Ferreira DG, Temido-Ferreira M, Vicente Miranda H, Batalha VL, Coelho JE, Szego EM, et al. Alpha-synuclein interacts with PrP(C) to induce cognitive impairment through mGluR5 and NMDAR2B. Nat Neurosci. 2017;20(11):1569–79.PubMedCrossRef Ferreira DG, Temido-Ferreira M, Vicente Miranda H, Batalha VL, Coelho JE, Szego EM, et al. Alpha-synuclein interacts with PrP(C) to induce cognitive impairment through mGluR5 and NMDAR2B. Nat Neurosci. 2017;20(11):1569–79.PubMedCrossRef
92.
Zurück zum Zitat Aulic S, Masperone L, Narkiewicz J, Isopi E, Bistaffa E, Ambrosetti E, et al. alpha-Synuclein Amyloids Hijack Prion Protein to Gain Cell Entry, Facilitate Cell-to-Cell Spreading and Block Prion Replication. Sci Rep. 2017;7(1):10050.PubMedPubMedCentralCrossRef Aulic S, Masperone L, Narkiewicz J, Isopi E, Bistaffa E, Ambrosetti E, et al. alpha-Synuclein Amyloids Hijack Prion Protein to Gain Cell Entry, Facilitate Cell-to-Cell Spreading and Block Prion Replication. Sci Rep. 2017;7(1):10050.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Haik S, Privat N, Adjou KT, Sazdovitch V, Dormont D, Duyckaerts C, et al. Alpha-synuclein-immunoreactive deposits in human and animal prion diseases. Acta Neuropathol. 2002;103(5):516–20.PubMedCrossRef Haik S, Privat N, Adjou KT, Sazdovitch V, Dormont D, Duyckaerts C, et al. Alpha-synuclein-immunoreactive deposits in human and animal prion diseases. Acta Neuropathol. 2002;103(5):516–20.PubMedCrossRef
95.
Zurück zum Zitat Gelpi E, Colom-Cadena M. Oligomers: a hot topic for neurodegeneration and a note of caution for experimental models. Brain. 2019;142(2):228–30.PubMedCrossRef Gelpi E, Colom-Cadena M. Oligomers: a hot topic for neurodegeneration and a note of caution for experimental models. Brain. 2019;142(2):228–30.PubMedCrossRef
96.
Zurück zum Zitat Katorcha E, Makarava N, Lee YJ, Lindberg I, Monteiro MJ, Kovacs GG, et al. Cross-seeding of prions by aggregated alpha-synuclein leads to transmissible spongiform encephalopathy. PLoS Pathog. 2017;13(8):e1006563.PubMedPubMedCentralCrossRef Katorcha E, Makarava N, Lee YJ, Lindberg I, Monteiro MJ, Kovacs GG, et al. Cross-seeding of prions by aggregated alpha-synuclein leads to transmissible spongiform encephalopathy. PLoS Pathog. 2017;13(8):e1006563.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat 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. Neurobiol Aging. 2003;24(2):197–211.PubMedCrossRef 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. Neurobiol Aging. 2003;24(2):197–211.PubMedCrossRef
98.
Zurück zum Zitat Braak H, Alafuzoff I, Arzberger T, Kretzschmar H, Del Tredici K. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol. 2006;112(4):389–404.PubMedPubMedCentralCrossRef Braak H, Alafuzoff I, Arzberger T, Kretzschmar H, Del Tredici K. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol. 2006;112(4):389–404.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Braak H, Braak E. Staging of Alzheimer's disease-related neurofibrillary changes. Neurobiol Aging. 1995;16(3):271–8 discussion 8-84.PubMedCrossRef Braak H, Braak E. Staging of Alzheimer's disease-related neurofibrillary changes. Neurobiol Aging. 1995;16(3):271–8 discussion 8-84.PubMedCrossRef
100.
Zurück zum Zitat Braak H, Thal DR, Ghebremedhin E, Del Tredici K. Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J Neuropathol Exp Neurol. 2011;70(11):960–9.PubMedCrossRef Braak H, Thal DR, Ghebremedhin E, Del Tredici K. Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J Neuropathol Exp Neurol. 2011;70(11):960–9.PubMedCrossRef
101.
Zurück zum Zitat Haroutunian V, Purohit DP, Perl DP, Marin D, Khan K, Lantz M, et al. Neurofibrillary tangles in nondemented elderly subjects and mild Alzheimer disease. Arch Neurol. 1999;56(6):713–8.PubMedCrossRef Haroutunian V, Purohit DP, Perl DP, Marin D, Khan K, Lantz M, et al. Neurofibrillary tangles in nondemented elderly subjects and mild Alzheimer disease. Arch Neurol. 1999;56(6):713–8.PubMedCrossRef
102.
Zurück zum Zitat Thal DR, Rub U, Orantes M, Braak H. Phases of a beta-deposition in the human brain and its relevance for the development of AD. Neurology. 2002;58(12):1791–800.PubMedCrossRef Thal DR, Rub U, Orantes M, Braak H. Phases of a beta-deposition in the human brain and its relevance for the development of AD. Neurology. 2002;58(12):1791–800.PubMedCrossRef
103.
Zurück zum Zitat Kovacs GG, Botond G, Budka H. Protein coding of neurodegenerative dementias: the neuropathological basis of biomarker diagnostics. Acta Neuropathol. 2010;119(4):389–408.PubMedCrossRef Kovacs GG, Botond G, Budka H. Protein coding of neurodegenerative dementias: the neuropathological basis of biomarker diagnostics. Acta Neuropathol. 2010;119(4):389–408.PubMedCrossRef
105.
Zurück zum Zitat Lue LF, Walker DG, Adler CH, Shill H, Tran H, Akiyama H, et al. Biochemical increase in phosphorylated alpha-synuclein precedes histopathology of Lewy-type synucleinopathies. Brain Pathol. 2012;22(6):745–56.PubMedPubMedCentralCrossRef Lue LF, Walker DG, Adler CH, Shill H, Tran H, Akiyama H, et al. Biochemical increase in phosphorylated alpha-synuclein precedes histopathology of Lewy-type synucleinopathies. Brain Pathol. 2012;22(6):745–56.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Muntane G, Ferrer I, Martinez-Vicente M. Alpha-synuclein phosphorylation and truncation are normal events in the adult human brain. Neuroscience. 2012;200:106–19.PubMedCrossRef Muntane G, Ferrer I, Martinez-Vicente M. Alpha-synuclein phosphorylation and truncation are normal events in the adult human brain. Neuroscience. 2012;200:106–19.PubMedCrossRef
107.
Zurück zum Zitat Larson ME, Sherman MA, Greimel S, Kuskowski M, Schneider JA, Bennett DA, et al. Soluble alpha-synuclein is a novel modulator of Alzheimer's disease pathophysiology. J Neurosci. 2012;32(30):10253–66.PubMedPubMedCentralCrossRef Larson ME, Sherman MA, Greimel S, Kuskowski M, Schneider JA, Bennett DA, et al. Soluble alpha-synuclein is a novel modulator of Alzheimer's disease pathophysiology. J Neurosci. 2012;32(30):10253–66.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Nemani VM, Lu W, Berge V, Nakamura K, Onoa B, Lee MK, et al. Increased expression of alpha-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron. 2010;65(1):66–79.PubMedPubMedCentralCrossRef Nemani VM, Lu W, Berge V, Nakamura K, Onoa B, Lee MK, et al. Increased expression of alpha-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron. 2010;65(1):66–79.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Lee MK, Stirling W, Xu Y, Xu X, Qui D, Mandir AS, et al. Human alpha-synuclein-harboring familial Parkinson's disease-linked ala-53 --> Thr mutation causes neurodegenerative disease with alpha-synuclein aggregation in transgenic mice. Proc Natl Acad Sci U S A. 2002;99(13):8968–73.PubMedPubMedCentralCrossRef Lee MK, Stirling W, Xu Y, Xu X, Qui D, Mandir AS, et al. Human alpha-synuclein-harboring familial Parkinson's disease-linked ala-53 --> Thr mutation causes neurodegenerative disease with alpha-synuclein aggregation in transgenic mice. Proc Natl Acad Sci U S A. 2002;99(13):8968–73.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Scott DA, Tabarean I, Tang Y, Cartier A, Masliah E, Roy S. A pathologic cascade leading to synaptic dysfunction in alpha-synuclein-induced neurodegeneration. J Neurosci. 2010;30(24):8083–95.PubMedPubMedCentralCrossRef Scott DA, Tabarean I, Tang Y, Cartier A, Masliah E, Roy S. A pathologic cascade leading to synaptic dysfunction in alpha-synuclein-induced neurodegeneration. J Neurosci. 2010;30(24):8083–95.PubMedPubMedCentralCrossRef
Metadaten
Titel
Beyond the synucleinopathies: alpha synuclein as a driving force in neurodegenerative comorbidities
verfasst von
Naomi P. Visanji
Anthony E. Lang
Gabor G. Kovacs
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Translational Neurodegeneration / Ausgabe 1/2019
Elektronische ISSN: 2047-9158
DOI
https://doi.org/10.1186/s40035-019-0172-x

Weitere Artikel der Ausgabe 1/2019

Translational Neurodegeneration 1/2019 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Demenzkranke durch Antipsychotika vielfach gefährdet

23.04.2024 Demenz Nachrichten

Wenn Demenzkranke aufgrund von Symptomen wie Agitation oder Aggressivität mit Antipsychotika behandelt werden, sind damit offenbar noch mehr Risiken verbunden als bislang angenommen.