Skip to main content

Neuropathology of incidental Lewy body & prodromal Parkinson’s disease

Abstract

Background

Parkinson’s disease (PD) is a progressive neurodegenerative disorder associated with a loss of dopaminergic (DA) neurons. Despite symptomatic therapies, there is currently no disease-modifying treatment to halt neuronal loss in PD. A major hurdle for developing and testing such curative therapies results from the fact that most DA neurons are already lost at the time of the clinical diagnosis, rendering them inaccessible to therapy. Understanding the early pathological changes that precede Lewy body pathology (LBP) and cell loss in PD will likely support the identification of novel diagnostic and therapeutic strategies and help to differentiate LBP-dependent and -independent alterations. Several previous studies identified such specific molecular and cellular changes that occur prior to the appearance of Lewy bodies (LBs) in DA neurons, but a concise map of such early disease events is currently missing.

Methods

Here, we conducted a literature review to identify and discuss the results of previous studies that investigated cases with incidental Lewy body disease (iLBD), a presumed pathological precursor of PD.

Results

Collectively, our review demonstrates numerous cellular and molecular neuropathological changes occurring prior to the appearance of LBs in DA neurons.

Conclusions

Our review provides the reader with a summary of early pathological events in PD that may support the identification of novel therapeutic and diagnostic targets and aid to the development of disease-modifying strategies in PD.

Pathology progression and staging in PD

The traditional concept of pathology progression in PD

Parkinson’s disease (PD) is the most common neurodegenerative movement disorder and is characterised by the progressive development of bradykinesia, muscular rigidity, rest tremor, and postural instability [1]. The cardinal motor features result from the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc) [2]. A neuropathological hallmark of PD are neuronal protein aggregates termed Lewy bodies (LBs) (Fig. 1a) [3]. LBs are composed of vesicular membrane structures and dysmorphic organelles in conjunction with protein aggregates containing alpha-Synuclein as the main component (αSyn) [4, 5]. Gene multiplications and missense mutations in SNCA, the gene coding for αSyn, are causative for familial forms of PD, which account for 10–15% of cases [6,7,8,9]. In addition, genome-wide association studies linked common variants at the SNCA locus to sporadic PD, thus further supporting an important pathogenic role of α-Syn in PD [10]. Postmortem studies suggested that the gradual appearance of LBs correlates with disease progression in PD [11]. Based on the gradual appearance of LBs, Braak et al. developed a neuropathological staging scheme for PD. The authors proposed that PD primarily starts in the olfactory bulb and the autonomic enteric nervous system (ENS), with a caudo-rostral (retrograde) spread of Lewy body pathology (LBP) over time, ultimately reaching the SNc where it is suspected to initiate the demise of DA neurons (Fig. 2) [12,13,14]. LBs are therefore considered to be a marker for disease progression, while neuronal loss represents a well-established neuropathological correlate of clinical PD (cPD) symptoms [3, 15, 16].

Fig. 1
figure 1

Neuropathological hallmarks of Parkinson’s disease and associated co-pathologies. a The neuropathological hallmarks of PD are shown in the three sub-panels. From left to right: a normal substantia nigra without any PD features; substantia nigra tissue from a PD patient with Lewy bodies (arrows) and pigmented/dopaminergic neuron loss; an immunohistochemical stain against αSyn (clone 42) from the same PD patient demonstrating intraneuronal Lewy bodies and Lewy neurites. Note that melanin pigment and DAB stain are not easy to distinguish. Scale bar = 50 μm. b Immunohistochemically stained sections of the amygdala from a patient suffering from mixed-type dementia that presented clinically as PD. The amygdala is a region commonly affected by co-pathologies. From left to right: An antibody against phosphorylated tau protein (clone AT8) demonstrates neurofibrillary tangles; immunohistochemistry against amyloid beta (clone 4G8) highlights diffuse and cored amyloid beta plaques; the clone 42 against αSyn marks Lewy bodies, Lewy neurites and a few axonal spheroids. Scale bar = 100 μm

Fig. 2
figure 2

Schematic illustrating disease progression from the pre-clinical phase of PD to the late clinical phase, including the associated clinical symptoms, Substantia nigra pathology and spread of Lewy body pathology according to Braak’s staging model. Note the presence of neuronal dysfunction in the SNc prior to the appearance of Lewy bodies in this area. Created with Biorender.com

Braak’s staging is divided into six different stages that reflect the progression of LBP from the dorsal motor nucleus of the vagus nerve (DMV) (Braak stage 1) to the locus coeruleus (LC) (Braak stage 2), SNc and amygdala (Braak stage 3) and ultimately reaching cortical areas (Braak stage 4–6) [17, 18]. The Braak stages correspond to the type and degree of clinical symptoms associated with disease progression. Early stages are characterised by non-motor symptoms, while typical PD motor signs are thought to appear once the SNc is affected at Braak stages ≥ 3, and cognitive symptoms arise only as LBP reaches the cortex in Braak stages 5 and 6 (Fig. 2). Braak’s model proposes that LBP gradually appears in defined anatomical structures during disease progression [3, 19]. In line with the idea of a prion-like LBP ‘spreading’ mechanism, fetal DA neurons transplanted into the PD SNc exhibited proteinaceous inclusions that resembled LBs [20, 21]. This result was interpreted as a ‘spread’ of LBP from the host to the graft. In mice, synthetic pre-formed αSyn fibrils propagate from the site of stereotaxic injection to synaptically connected, neighboring structures, thereby creating a Lewy-like pathology [22,23,24,25]. Similarly, proteins extracted from human brains with LBP and injected into the striatum of monkeys can also propagate to neighboring structures, illustrating LBP's propensity to spread from its origin [26, 27]. In summary, previous results support a model where LBP gradually builds up in clearly defined brain regions, leading to neuronal death. This process occurs according to a well-defined pattern, resulting in some brain areas remaining unaffected until the final stages of PD, whereas others are devastated by degeneration early on.

Controversies and limitations of the braak staging scheme

The co-incidence of DA neuron loss and LBs initially primed the conclusion that these intraneuronal inclusions—in combination with cell death—were responsible for the disease. However, a considerable body of research appears to give rise to concerns regarding the overall significance of LBP. For instance, Gibb et al. reported an age-dependent increase in the prevalence of LBs from 3.8% to 12.8% between the sixth and ninth decades of age. This amount exceeds the prevalence of PD by about three- to six-fold [28, 29]. In line, previous reports found that a significant proportion of neuropathologically confirmed LBD cases never exhibited clinical symptoms [30]. In addition, more recent research demonstrated that cell death and LBP do not entirely correlate in the affected brain regions. For instance, even in the absence of LBP, there is considerable neuronal death in the supraoptic nucleus in PD. By contrast, there is no discernible neuron loss in the neighboring, LB-rich tuberomammillary nucleus of the hypothalamus [31]. Furthermore, in patients who do not exhibit dementia during disease progression, the only cortical region that shows substantial neuronal loss is the pre-supplementary motor cortex, where small intra-telencephalic pyramidal neurons degenerate in the absence of LBP [32, 33]. These results cast doubt on the concept that cell death is a consequence of LBP in PD.

Conflicting with other reports, which claim that neurons are primarily lost in brain regions with LBP [34,35,36], a study from Iacono et al. found no significant correlation between neuronal loss and LBs in the PD brain [37]. Another line of evidence for an LBP-independent pathological process comes from PD cases carrying genetic mutations where LBP distribution is distinct from that of idiopathic PD [38]. For instance, only a part of PD patients with a G2019S mutation in the LRRK2 gene exhibit LBP and most patients with other LRRK2 mutations do not even show LBP at all [39] despite substantial SNc DA neuronal degeneration. Likewise, PD cases with PARK2 mutations have only sparsely distributed LBP with a pattern distinct from that found in idiopathic PD cases [40], although these cases may be phenotypic variations. Finally, conflicting evidence comes from neuropathological and histological studies. Because the number of LBs in patients with mild to moderate SNc neuron loss was higher than in patients with severe neuronal depletion, LB-containing neurons have been initially assumed to be the dying neurons [41]. However, Tompkins and Hill demonstrated that the presence of LBs does not predict a higher degree of cell death compared to the general population of SNc neurons and that most neurons that undergo cell death do not even contain LBs [42].

Moreover, whether SNc neurons contain LBs or not, they are similarly affected by morphological dendritic abnormalities or biochemical changes, indicating that DA neurons, in general, are involved in a yet-to-be-defined disease process [43,44,45,46]. These results imply that region-specific environmental changes may prime these DA neurons to preferentially degenerate in PD. Consequently, attempts to correlate the density of either cortical or brain stem LBs with the progression and severity of clinical PD symptoms were unsuccessful [47,48,49,50]. Along these lines, in a certain percentage of PD patients who developed dementia, no LBs could be detected in cortical areas or other areas outside the brain stem [51, 52], and these cases may suffer from concomitant amyloid beta (Aβ) pathology. Conversely, the simultaneous presence of Lewy body pathology and Alzheimer's Disease (AD)-related changes, such as hyperphosphorylated tau protein or Aβ, can also be observed (Fig. 1b). Conversely, LBP, which is typically found in the amygdala, is frequently detected in AD cases [53].

Collectively, these findings indicate that the pathophysiology of neurodegeneration and cell death can hardly be explained by LBs or LBP-related cell death alone. Alternative views are that LB formation is a process for detoxification of pathological αSyn aggregates located at a harmful site in the neuron, such as the presynapse [54]. Studies investigating the ultrastructure of LBs indicated that they are formed in an aggresome-related process and support the notion that LBs are a way of containment of protein aggregates and degraded organelles [5, 55]. By using correlative light and electron microscopy and tomography on postmortem human brain tissue from PD brains, the study done by Shahmoradian et al. found a crowded environment of membranes in LBs, including vesicular structures and dysmorphic organelles [5]. Crowding of organellar components was confirmed by stimulated emission depletion (STED)-based super-resolution microscopy, and a high lipid content within LBs was corroborated by confocal imaging, Fourier-transform coherent anti-Stokes Raman scattering infrared imaging and lipidomics. The latter report suggests that lipid membrane fragments and distorted organelles, together with a non-fibrillar form of αSyn, are the main structural components of LBs and that they do not contain fibrillar αSyn aggregates. Although a matter of current debate [56], these results thus challenge the pivotal role of αSyn and point towards cellular and molecular changes that may occur independent from the formation of LBP. This view is supported by studies implicating mutations in PINK1 [57] and lysosomal genes in PD, such as GBA [58,59,60,61]. Conversely, out of 2000 PD cases, only 0.05% had mutations in the SNCA gene, creating uncertainty about how αSyn accumulates in the other 99.95% of cases and whether the protein has a causative role in PD [62]. In summary these results thus cast doubt on the significance of LBP as the sole disease-causing factor in PD and alternative models are required to explain such apparent discrepancies.

Investigating pathological precursors to determine the significance of LBP in PD

A small number of conceptual approaches attempted to dissect LBP-dependent from LBP-independent events in the PD brain. Besides comparing LBP-affected and unaffected neurons, one strategy has been the examination of pathological precursors of LBP. In addition to cPD, there are a few cases that exhibit brain stem-restricted LBP in the absence of the characteristic clinical PD symptoms and these cases are referred to as incidental Lewy body disease (iLBD) [14, 63, 64]. iLBD occurs in 10–15% of people over 60 years of age [65] and is assumed to represent a pathological precursor in PD [66,67,68,69]. Whether these cases constitute a neuropathological PD precursor has been a matter of controversy for some years, as there is no proof that these cases would have progressed to PD if they had survived longer or instead simply recapitulated features of normal brain ageing [70]. However, the study of consecutive cases in large case series and the recognition of several intermediate degrees of involvement of the brain stem, limbic structures and, eventually, the cerebral cortex supports the argument for iLBD as a pathological precursor stage of PD. Since SNc neurons are, by definition, still spared from LBs in iLBD, investigating SNc neurons in these cases may provide insight into the cellular and molecular changes occurring at this critical site in the absence of LBP [71], thus allowing to distinguish LBP-dependent from LBP-independent changes. Moreover, investigating SNc neurons in iLBD may support the understanding of early pathological events occurring prior to the appearance of LBP. Because therapeutic approaches that delay or slow down disease progression in PD are likely to be more effective prior to neuronal cell death, the examination of SNc neuronal changes in iLBD cases may aid to the identification of novel therapeutic targets and, ultimately, to the development of early-acting, potentially disease-modifying interventions. Based on these two reasons, we believe that investigating SNc neuronal changes in iLBD warrants further research. Here, we will review the evidence available from earlier studies that examined the molecular and cellular changes in the iLBD SNc to create a concise map of early neuropathological events during PD disease progression and to nurture prospective research in this direction.

Neuropathology of incidental LBD (iLBD)

Structural changes in iLBD

Neuropathological studies suggest that by the time a patient is diagnosed with PD based on clinical motor symptoms, a significant proportion of DA neurons is already lost (Fig. 2) [72] and within four years of diagnosis, DA terminals in the dorsal putamen almost entirely disappeared [73]. Therefore, estimates propose that at least cell loss in the brain commences 5 to 10 years prior to the clinical diagnosis [74, 75]. Indeed, several previous studies investigating neuropathological changes in iLBD demonstrated structural changes in SNc DA neurons in the absence of LBs (Table 1, Fig. 2). In accord with an early neuronal malfunction, previous studies demonstrated a substantial (10–20%) loss of SNc DA neurons and impaired nigrostriatal integrity at Braak stages 1 and 2 [76,77,78]. Likewise, Dijkstra et al. found a 20% decrease in SNc neuronal cell density in iLBD compared with controls [3]. More recently, Iakono et al. demonstrated a marked nigral neuronal loss in PD and iLBD compared to control cases [37]. Milber et al. have shown that neuronal dysfunction and cell loss may precede LBP in the SNc because prior to the appearance of LBs, these processes were observed in the SNc in iLBD at comparable levels to those of higher Braak stages [79]. In accord with a functionally relevant disease process occurring prior to LBP, PD motor symptoms have even been reported at stage 2 of Braak [80]. All these results are also further supported by case reports [81]. These findings illustrate the need for further investigation at these early stages to account for the neuronal loss before the onset of LBP in this area.

Table 1 Table summarizing structural and functional changes in the iLBD midbrain

General neurochemical changes in iLBD

In addition to these structural changes (i.e., cell loss), investigating iLBD brains revealed certain neurochemical alterations. For instance, Dickson et al. found that tyrosine hydroxylase (TH) immunoreactivity in the striatum was decreased in iLBD compared to normal controls, but not to the same extent as in PD [71]. TH is an enzyme critical for DA production, and its decrease in iLBD indicates a nigrostriatal system that is already impaired at this early stage. Using quantitative ELISA, Beach et al. demonstrated that striatal TH showed a 49.8% reduction in iLBD cases compared to control cases [82]. Together with the morphological studies described above, these reports suggest an early neurochemical alteration of SNc DA neurons prior to the appearance of LBs. Other research groups have provided additional findings on the early pathological changes in PD, including neurochemical or metabolic changes. For instance, early oxidative damage was found in the SNc in iLBD, where nitrated αSyn is already present in small granules in DA neurons before the appearance of LBs [83]. The authors thus concluded that oxidative damage is an early event in PD and may precede the formation of LBs.

In the context of the Renin-angiotensin system (RAS), it is intriguing to note that although this hormonal system is traditionally associated with regulating blood pressure, there is significant interplay with the DA system [112, 113]. Studies have demonstrated that angiotensin blockers can exert a neuroprotective effect on midbrain DA neurons both in vivo and in vitro by reducing oxidative stress, thereby indicating their potential as a therapeutic option. For instance, a retrospective study focusing on patients receiving angiotensin blockers as treatment for hypertension showed a reduced risk of developing PD [114]. Similarly, an analysis of data from ischemic heart disease patients revealed that those prescribed with angiotensin II inhibitors—which have the capacity to cross the blood–brain barrier—had a lower risk of developing PD [115]. These findings underscore the potential of these compounds to counteract the early oxidative damage that primes DA neurons for degeneration, thereby presenting a promising strategy for reducing PD risk.

Further biochemical studies have shown increased levels of neuroketals in the SNc in post mortem tissue from Braak stages 1 and 2, supporting the notion that oxidative damage to specific lipids in the SNc occurs at very early stages of PD and prior to the appearance of LBP [83, 84]. In line, recent observations have shown the concentration of L-ferritin in the SNc to be lower in iLBD (and PD) compared with controls, whereas H-ferritin in PD was found to be higher than in iLBD and controls. This illustrates the subtle abnormalities in iron metabolism in the SNc at the early stages of PD [85]. Summarising these results, neurochemical changes occurring prior to LBP may contribute to the increased propensity of SNc DA neurons to degenerate.

Changes in autophagy

In line with these neurochemical changes, a report demonstrated p62 immunoreactivity in association with abnormal αSyn inclusions at the early stages of LBP, thus suggesting premature alterations to autophagic pathways in these cases [87]. Tang et al. recently investigated autophagy-associated SNARE molecules in post mortem brain tissue from LBD cases and found a stage-dependent decline of the v-SNARE SNAP29 – a member of the SNARE complex mediating autophagolysosome fusion – as early as in Braak stage 1 (Table 1) [86]. Additional experiments in cultured dopaminergic neurons demonstrated αSyn overexpression to reduce autophagy turnover by compromising the fusion of autophagosomes with lysosomes, thus leading to a decrease in the formation of autophagolysosomes. Mechanistically, αSyn interacted with and decreased the abundance of SNAP29 in vitro. Furthermore, SNAP29 knockdown mimicked the effect of αSyn on autophagy, whereas SNAP29 co-expression reversed the αSyn-induced changes on autophagy turnover and ameliorated DA neuronal cell death. These results thus demonstrated a previously unknown capacity of αSyn to affect intracellular autophagy-associated SNARE proteins and, consequently, reduce autophagolysosome fusion. Most notably, this effect may be evident before the presence of LBs in the SNc. Whereas SNAP29 loss has been identified in SNc neurons in iLBD, the cell culture work is derived from αSyn over-expression, thus making it difficult to compare the two results. One possible explanation is that oligomeric αSyn, not yet aggregated into LBs, may cause such cellular changes during early pathology, although specific αSyn-species remain to be identified.

Oligomers, which are small aggregates of misfolded proteins, are believed by some to be a key contributor to the neurodegenerative processes that occur in PD [116]. These oligomers are thought to be more toxic than other forms of αSyn, such as monomers or fibrils, and have been shown to impair the function of neurons in cell culture and animal models of PD. Furthermore, recent research has indicated that αSyn oligomers can spread from cell to cell in a prion-like manner, propagating the disease throughout the brain [117]. This has led to the hypothesis that targeting αSyn oligomers could be a promising therapeutic strategy for PD. Whereas LBP is visible with histologic methods αSyn oligomers remain undetectable with routine approaches but may be an important contributor of early pathological changes. Detecting αSyn oligomers requires special techniques, and their distribution and association with clinical features are important research objectives. Recent advances in detecting αSyn oligomers, such as using proximity ligation assay (PLA) [118] or oligomer-specific antibodies [119] may support investigating such early pathological changes in PD.

Immunological changes in iLBD

Following clinical reports [120], a recent immunohistochemical study assessing the abundance of the inflammation-associated Toll-like-Receptor 2 (TLR-2) showed increased numbers of TLR-2-positive microglia in the iLBD SNc compared to PD [88], suggesting inflammatory changes occur at early stages and prior to the development of PD symptoms. By contrast, there was a progressive increase from control to PD in the numbers of CD68-positive microglia/macrophages, a marker associated with phagocytosis, although an increase in the number of microglia was not identified [89]. Walker et al. examined the differential expression of inflammatory and trophic molecules in the SNc and striatum of control, iLBD and PD cases and found distinct patterns of inflammation and growth factor changes [91], which was also reinforced by animal studies [121]. Another piece of evidence suggesting early immunological changes came from the work of Galioano-Landeira et al. The authors found that CD8-positive T-lymphocytes were increased in the SNc of PD cases compared to the control group, whereas CD4-positive T cells remained unchanged [92]. Most notably, a robust infiltration of CD8-positive T-cells has been observed prior to the appearance of LBP (Braak Stage 1) and in the absence of DA cell death. CD8-positive T-cells were found to be equipped with cytolytic enzymes (granzymes A, B and K) and proinflammatory cytokines (interferon gamma) with phenotypic differences between early and late stages. A high proportion of nigral CD8 T cells were identified as tissue-resident memory T cells. These results identified a substantial nigral cytotoxic CD8-T-cell infiltration as an early pathogenic event preceding LBP and DA cell death in PD. This further highlights microenvironmental changes which may impact later nigral cell survival. In another study by Hurley et al., iLBD cases had an increased number of IBA1-positive microglia. In the anterior cingulate cortex (ACC), PAR2-positive microglia were increased in iLBD, while in the primary motor cortex, tyrosin-1 was increased in microglia. However, TH-positive neurons in the SNc only showed a decreasing trend [90]. Doorn et al. investigated microglia activity by quantifying the minichromosome maintenance protein 2 (MCM2), a cell proliferation marker. The authors found MCM2-positive cells to be increased in the hippocampus (HC) of iLBD cases but not in established PD patients. This study thus suggests an early microglial response in the HC, indicating that neuroinflammatory processes play an essential role in developing PD pathology [88]. Finally, in another study, the tissue from different Braak stages was examined for the presence of integrin αvβ3, a marker for angiogenesis, along with vessel number and activated microglia. In this study, all PD cases had greater levels of αvβ3 in the SNc compared to controls. PD subjects also had increases in microglia number and activation in the SNc, suggesting a link between inflammation and clinical disease, whereas microglia activation in iLBD subjects was limited to the LC, an area involved in early-stage PD [89]. In summary, immune-associated changes appear to occur early during disease progression, and, consequently, anti-inflammatory strategies may be potentially disease-modifying for PD. Indeed, several anti-inflammatory drugs have been tested for their therapeutic potential in PD. For instance, statins have been proposed to exert neuroprotective effects in PD models through an anti-inflammatory response, improving motor function and attenuating the increase in inflammatory cytokines. Simvastatin, for example, effectively crosses the blood–brain barrier and is currently being studied in a phase 2 randomized, placebo-controlled futility trial [122]. Although recently announced results indicated futility for slowing the progression of PD, an anti-inflammatory approach may require early treatment before LBP-related cell death to yield successful therapeutic effects [123,124,125]. Other clinical trials investigating anti-inflammatory agents are also still ongoing [126, 127].

Early synaptic pathology in LBD

Mounting evidence indicates that SNc DA neuron degeneration is likely to start from synaptic pathology [128] and that the loss of synaptic connectivity may precede nerve cell loss. As early as 1989, by analysing vesicular monoamine transporter 2 (VMAT2) binding during ageing in PD and healthy subjects, Sherman et al. provided the first evidence indicating that PD symptoms appear when the striatal denervation state is over a critical threshold of about 50% [82, 93, 94]. This illustrated the relevance of synaptic terminal degeneration in the onset of disease and its clinical phenotype [129]. Schulz-Schaeffer et al. reported that αSyn pathology mainly involves synaptic compartments and proposed that the first neuronal compartment affected by its deposition might be the synaptic terminal [95]. In accord with an early synaptic pathology in PD, most αSyn aggregates accumulated at presynaptic terminals in paraffin-embedded tissue blots from LBP cases [130]. Thus, at the onset of clinical motor symptoms, the loss of DA synaptic terminals exceeds the loss of DA cell bodies, pointing towards an early alteration of synaptic projections that precede neuronal death.

Moreover, neuroanatomical studies of post mortem brain samples from familial PD cases support the idea that synaptic decay precedes neuronal death [131, 132]. These observations support a ‘dying back’ hypothesis where synaptic demise, including presynaptic dysfunction, occurs prior to neuronal death [133, 134]. This view is supported by a series of preclinical studies indicating that αSyn aggregation at synaptic sites impairs neuronal function and axonal transport by affecting synaptic vesicle release [135]. Numerous studies found pre- and postsynaptic structural integrity alterations in PD and Dementia with Lewy bodies (DLB) [46, 130, 136,137,138,139]. Furthermore, apart from αSyn, several other PD-associated proteins such as leucine-rich repeat kinase 2 (LRRK2), parkin, DJ-1, PINK1, Rab38B and synaptojanin have been found to be involved in the control of DA synaptic function [140,141,142,143,144,145]. In accord with an early synaptic dysfunction in PD, various in vivo imaging studies demonstrated presynaptic neurotransmitter deficiencies in PD [136]. These findings seem to indicate that the degenerative process in PD is – at least in part – located at the presynapse, ultimately resulting in a neurotransmitter deficiency syndrome [146, 147]. This degeneration of synapses appears to emerge before motor symptom onset; however, the exact timeline of this progression and its clinical correlates are yet to be fully elucidated. Another critical aspect of these studies is that none of such results were derived directly from iLBD cases, and, although it is conceivable that, for instance, oligomeric non-aggregated αSyn species affect synaptic function prior to the appearance of typical LBs, the specific significance of such αSyn species remains uncertain.

Changes in gene expression & cell types

A relevant study on early transcriptomic changes in PD was conducted by Wilma van den Berg's group using RNA microarrays [96]. The authors aimed to elucidate molecular mechanisms underlying neuronal dysfunction and LBP in the pre-motor phase of PD and investigated the transcriptome of the SNc of well-characterised iLBD, PD and age-matched controls. Before SNc-LBP, at Braak stages 1-2, they observed deregulation of pathways linked to axonal degeneration, immune response, and endocytosis, including axonal guidance signalling, mTOR signalling, eIF2 signalling and clathrin-mediated endocytosis in the SNc. The results indicate molecular mechanisms related to axonal dysfunction, endocytosis and immune response are already affected before LBP reaches the SNc, while mTOR and eIF2 signalling is also impaired during later stages.

Interesting work implicating additional cell types in iLBD came from a study that integrated genome-wide association study results with single-cell transcriptomic data from the entire mouse nervous system to systematically identify cell types underlying brain complex traits [98]. When applying expression-weighted cell-type enrichment (EWCE) to data from previous studies [148, 149], the authors found that downregulated genes in PD were enriched explicitly in DA neurons (consistent with the loss of this particular cell type in the disease). In contrast, upregulated genes were significantly enriched in cells from the oligodendrocyte lineage. When analysing gene expression data from post mortem human brains, downregulated genes were not enriched in DA neurons at Braak stage 1–2. Conversely, upregulated genes were already strongly enriched in oligodendrocytes at this stage, thus indicating that their involvement precedes the emergence of pathological changes in the SNc. In summary, this study thus supports an early alteration of oligodendrocytes preceding LBP in PD, although the data were in part based on investigating mice.

This finding was corroborated by a recent single-cell study where significant associations were found between reported PD risk genes and highly expressed genes in oligodendrocytes. Furthermore, the risk for PD age of onset was associated with genes highly expressed in oligodendrocyte precursor cells [99]. These studies thus support an early alteration of oligodendrocytes and their precursors, preceding LBP in PD. A study by Santpere et al. investigated global transcriptional changes in the frontal cortex (Area 8) in iLBD, PD and DLB. The authors identified different co-expressed gene sets associated with disease stages. They conducted a functional annotation of iLBD-associated modules using the gene ontology framework categories enriched in gene modules and differentially expressed genes, including modules or gene clusters correlated to iLBD. These clusters revealed upregulated dynein genes and taste receptors and downregulated genes related to innate inflammation [100], thus demonstrating transcriptomic alterations in cortical brain areas in iLBD. In 2012, a study by Lin et al. investigated the extent of mtDNA mutations in early-stage PD and iLBD cases and found that mtDNA mutation levels in SNc neurons are significantly elevated in these cases [97]. However, this study defined iLBD by the absence of clinical parkinsonism or dementia but with Lewy bodies present in the SN, which corresponds to Braak stage 3. These findings illustrate the widespread transcriptomic changes preceding LBP, affecting various cell types, and deregulating crucial molecular pathways.

Proteomic changes

Changes in the expression of various additional proteins have also been demonstrated, for instance, by Wilhelmus et al., who reported an aberrant ApoE and low-density lipoprotein receptor-related protein 1 expression in SNc DA neurons in PD and iLBD cases. The authors concluded that alterations in lipoprotein homeostasis/signalling in DA neurons of the SNc constitute an early disease event during PD pathogenesis [101]. Likewise, changes in neuropeptides and glutathione levels were found in iLBD [102, 103]. Wilkinson identified changes in the glycosylation of proteins in iLBD: a total of 70 O-glycans were identified, with iLBD exhibiting significantly decreased levels of mannose-core and glucuronylated structures in the striatum and PD presenting an increase in sialylation and a decrease in sulfation [104]. Early oxidative damage in the frontal cortex of iLBD cases has been suggested by a study that investigated lipoxidation of the glycolysis-associated enzymes aldolase A, enolase 1, and glyceraldehyde dehydrogenase (GAPDH) [150] and likewise early work from Jenner et al. suggested a loss of glutathione (GSH) to be associated with iLBD [103, 105, 106]. These proteomic modifications furthermore exemplify the various changes in the SNc prior to LBP emergence.

Changes in neuronal function and excitability in iLBD

Changes in neuronal function and excitability may occur a long time before structural events can be appreciated and recent research began to elucidate the molecular factors governing such early neuronal malfunction. For instance, Tan et al. investigated the effect of αSyn on regulatory molecules in DA SNc neurons and found a loss of the Fragile X Mental Retardation Protein (FMRP) in most neuromelanin-positive neurons of the SNc in human post mortem brain tissue from PD and iLBD cases [107]. Because FMRP regulates the expression and function of numerous neuronal genes [151, 152], these results further suggest that in PD, DA neuron dysfunction is likely to be present long before morphological and histopathological changes and that the loss of FMRP in the SNc may be a key molecular event in these stages (Fig. 2). Loss of FMRP may have beneficial or detrimental effects on neuronal function in the SNc. Tan et al. demonstrated that the absence of FMRP ameliorates αSyn-induced DA dysfunction, and suggest that the early loss of FMRP in PD may in fact protective effects in PD. However, as with the aforementioned studies on autophagy, results from investigating αSyn over-expression are difficult to compare with human LBP and its sequential appearance as the specific αSyn species that are present at different time points are not yet known. The specific significance of FMRP for PD disease progression thus remains to be defined.

Peripheral changes in iLBD

In addition to these reported CNS changes, iLBD cases may exhibit both peripheral and autonomic pathological changes [153, 154]. For instance, a study by Beach et al. examined the presence of LBP in the gut of iLBD, PD and control cases. The authors found that in the vagus nerve, none of the healthy control subjects showed aggregates of phosphorylated αSyn (p-αSyn), while 46% of iLBD and 89% of PD cases were p-αSyn-positive. In the stomach, none of the control subjects had p-αSyn while 17% of iLBD and 81% of PD subjects did [108]. Following these findings, iLBD cases were retrospectively found to exhibit a lower frequency of bowel movements [109]. In a retrospective autopsy-based study of the human submandibular gland, PD and iLBD cases had LBP in the submandibular glands, the cervical superior ganglia, the cervical sympathetic trunk and vagal nerves [110]. Some previous work even suggested the presence of LBP in the spinal cord of iLBD cases [155] and another study, although limited by a small sample size, found a decrease of TH immunoreactivity within epi- and myocardial sympathetic nerve fibres in PD and iLBD [111]. These studies appear to confirm the cumulative results from studying prodromal PD (pPD), where αSyn is present in the peripheral and autonomic nervous system.

Prodromal PD as a clinical surrogate for early pathological changes

In addition to investigating iLBD, some previous studies investigated cases that exhibit so-called prodromal symptoms: prior to the appearance of the classic motor symptoms during cPD, most PD patients experience several typical non-motor signs that are collectively referred to as pPD (Fig. 3). These signs include REM sleep behaviour disorder (RBD), olfactory loss, constipation, autonomic dysfunction, psychiatric symptoms, and pathological imaging markers of the presynaptic dopaminergic and autonomic nervous system [156, 157]. These prodromal signs and symptoms often precede cPD by 10-20 years [158, 159]. As such, investigating pPD would contribute to understanding early pathological events in PD and indeed, studies that examined pPD have contributed some indirect evidence for early pathological changes, although these results were primarily derived from imaging results. For instance, MRI data from isolated RBD (iRBD) cases showed structural alterations in the SNc and grey matter changes in the motor cortico-subcortical loop correlated with motor abnormalities [160, 161]. iRBD is considered to be an early clinical sign during disease progression with a > 80% risk of conversion to cPD [162,163,164] within 15 years. Patients typically present with vivid, often frightening dreams that lead to vocalisation and sudden body movements (Fig. 3). In addition to these characteristic sleep disturbances, some iRBD cases may exhibit mild motor deficits [165,166,167,168] (Table 2). Such clinical data are consistent with an early affection of extrapyramidal motor areas during disease progression, although the specific molecular correlate remains uncertain. Furthermore, iRBD cases exhibit a reduced striatal dopamine transporter (DaT) binding [169, 170] on [123I] Ioflupan scintigraphy and an altered [18F]AV133 VMAT2 positron emission tomography (PET) signal [171], further indicating impaired integrity of the nigrostriatal pathway in these cases. Reduced DaT binding also seems to be correlated with changes in brain glucose metabolism as assessed by [18F] fluorodeoxyglucose ([18F]FDG) PET [172]. Likewise, iRBD cases exhibit impaired nigrostriatal connectivity as assessed by fMRI and ultrasound [161, 173,174,175,176,177,178,179,180,181,182,183] (rev. in [184]). In accord with the aforementioned pathological studies in iLBD, a study examining inflammatory changes in the SNc by [11C]PK11195 18 kDa translocator protein (TSPO) PET found increased microglial activation in iRBD, suggesting early immunological changes in the midbrain [120]. Furthermore, Imidazoline 2 imaging with [11C]BU99008 PET indicated activated astrocytes in early PD but even decreased tracer signal at late stages compared to healthy controls [185]. These imaging results thus collectively confirm an early and possibly inflammatory pathology in the PD midbrain. Overall, these observations thus indicate that in iRBD, the disease process extends beyond the sleep-related structures in the brainstem to other structures, including the nigrostriatal system [186, 187]. As a limitation of considering iRBD as pre-LBD cases, it is noteworthy that iRBD cases may exhibit LBP at different Braak stages, including those > 3, as substantiated by clinical findings [188] and that in some cases, iRBD may develop into Multiple System Atrophy (MSA) or Dementia with Lewy bodies (DLB) instead of cPD [164, 189].

Fig. 3
figure 3

Schematic summarizing clinical signs and symptoms of pPD and the approximate timescale for conversion to Clinical PD. Created with Biorender.com

Table 2 Table summarizing changes in the midbrain of pPD cases

In addition to these imaging results, several laboratory results have been derived from iRBD cases. For instance, serum neurofilament light chain (sNfL), a neuronal cytoskeletal protein released upon neuronal damage, might mark the conversion of iRBD to cPD [190]. Techniques such as proteomics analysis of serum samples have identified numerous proteins at significantly altered expression levels, providing further insight into the protein signature profile and molecular pathways involved in the pathogenesis of iRBD [51, 52]. In addition, alterations in circulating microRNAs (miRNAs) have been shown in iRBD. For instance, one study found miR-19b to be significantly down-regulated in iRBD cases that later converted to cPD but not in those who remained disease-free for several years, possibly indicating a role of miR-19b during early disease progression [194]. Still, the diagnostic value of serum miRNA detection remains controversial, as miRNAs show strong pleiotropy. For example, miR-19b has also been implicated in lung cancer progression [195] and schizophrenia [196]. One study revealed decreased antioxidant superoxide dismutase and increased glycolysis in iRBD cases using peripheral blood mononuclear cells [197]. The diagnostic value of other biospecimens, such as those from saliva, tears, or the microbiome, is yet to be explored in patients with iRBD, and longitudinal studies are required to establish whether such biosamples will support the understanding of disease onset and progression in LBP [198]. Finally, novel methods have been developed to investigate αSyn in iRBD cases by using Real-Time Quaking-Induced Conversion assays (RT-QuIC) [199,200,201]. These assays can detect αSyn seeding activity in different LB-associated conditions with a high sensitivity and specificity [202]. For instance, in a recent study that examined patients with iRBD, RT-QuIC detected misfolded α-Syn in the CSF with both sensitivity and specificity of 90%, and αSyn-positivity was associated with an increased risk of subsequent conversion to cPD or DLB [162]. Along these lines, another report aimed to detect of αSyn aggregates in the olfactory mucosa of a large cohort of subjects with iRBD by RT-QuIC [203]. The authors found the olfactory mucosa to be α-Syn-positive in 44.4% of iRBD cases, in 46.3% of cPD cases, but only in 10.2% of the control subjects. While the sensitivity for iRBD and cPD vs. controls was comparably low (45.2%), the specificity was found to be sufficiently high (89.8%). Compared to immunofluorescent techniques (IF) RT-QuIC was found to exhibit a high diagnostic accuracy [204].

In addition to iRBD, hyposmia is common in cPD (90%) and iRBD (67%) and sometimes precedes motor symptoms by > 20 years (Figs. 2 and 3) [158, 205,206,207]. The Prospective Validation of Risk Factors for the Development of Parkinson Syndromes (PRIPS) study found that cases of hyposmia had a fourfold risk of converting to cPD compared to normosmic cases [208]. An impaired sense of smell can thus be regarded as an early clinical event during disease progression [209, 210]. However, hyposmia alone is likely a suboptimal predictor for developing cPD since smell loss is relatively common in older adults, and only a minority will develop PD [211]. Concerning pathological changes in midbrain motor circuits, Sommer et al. identified 30 patients with idiopathic olfactory loss and found that 11 had increased echogenicity of the SNc on transcranial sonography and 5 cases had impaired DaT binding. This further supports early structural changes during the disease course [191]. Moreover, studies have shown a correlation between olfactory performance and DaT binding in early PD [192]. In another study, 11% of random hyposmic subjects had a DaT deficit at baseline compared to 1% of normosmic subjects [193]. Congruently with clinical studies on olfactory function, a study by Silveira-Moriyama et al. examining the post-mortem tissue from iLBD, PD and control patients found LBP in all samples from the olfactory bulb and the primary olfactory cortex in iLBD and PD cases [212]. Another study found that in both iLBD and PD tissue, the olfactory bulb was the region most frequently affected by LBP [19]. However, the immediate correlation between hyposmia and LBP in the olfactory bulb has yet to be substantiated since records regarding hyposmia in these patients studied were unavailable.

Collectively, results from investigating pPD further confirm an early structural and functional defect in motor-associated extrapyramidal circuits during PD disease progression that appears to be present prior to the evident appearance of motor signs and symptoms. On the downside, these results provide little conceptual insight into the mechanism of early midbrain neuron dysfunction in PD and no direct correlation with LBP.

Critical appraisal & future research directions

In the previous sections, we reviewed studies that collectively examined early pathological changes that precede the onset of LBP in the SNc. Investigating these changes has the potential to expose the significance of LBP, reveal early diagnostic and therapeutic targets and ultimately support the development of novel disease-modifying therapies for PD. However, all these approaches have conceptual shortcomings. Although investigating pPD cases by clinical and pathological methods supported the understanding of disease progression on a systemic level and generated valuable predictive data, it provided insufficient insight into the specific molecular and cellular changes occurring prior to LBP and cell death. This limitation applies particularly to areas in the brain stem and midbrain that are difficult to access in detail by routine diagnostics or tissue biopsies, including SNc DA neurons. Similar limitations apply to the neuropathological investigation of genetic PD cases [213], where genetic alterations (LRRK2, GBA, SNCA) predict the development of cPD prior to motor symptoms. Second, it is noteworthy that pPD cases may or may not exhibit LBP in SNc neurons, thus confounding the distinction between LBP-dependent and -independent changes. Although some iRBD cases may exhibit mild motor deficits [165,166,167,168], indicating SNc dysfunction, it is unclear if this is a consequence of LBP-associated cell degeneration or LBP-independent neuronal malfunction. Thus, investigating pPD does not truly help to clarify LBP's causative role in SNc DA neuron degeneration. Therefore, more research should focus on elucidating the relationship between these individual aspects of early disease events in PD and how they might correlate to one another.

A shortcoming of investigating iLBD relates to the uncertain progression pattern of LBP. Previous work suggested that only about 50% of all PD patients have a distribution of LBP in the brain that is entirely consistent with the Braak staging model, a prerequisite for the assumption that iLBD is a precursor for SNc pathology in PD [29, 214, 215], and about half of PD cases do not seem to show a caudo-rostral spread of LBP throughout the brain [216]. Furthermore, experimental evidence suggested that the spreading of αSyn via autonomic nerve fibres may occur in a caudo-rostral but also rostro-caudal direction [217,218,219]. In order to explain these distinct spreading patterns in PD, alternative ‘body-first’ and ‘brain-first’ models have been developed [157, 220,221,222]. As such, a brain-stem LBP would be the most common precursor of cPD, whereas a second route would commence in limbic areas, including the amygdala and progress to the SNc in a rostro-caudal spread. Although these theoretical models may partially explain the experimental inconsistencies, conclusions drawn from iLBD cases may be impeded by the uncertain correlation between clinical and neuropathological progression. Another concern regarding the Braak staging has finally been raised by earlier work form Schulz-Schaeffer et. al. These authors suggested that instead in the form somatic LBs, > 90% of αSyn aggregates are located at the presynapses in the form of very small deposits in PD, while postsynaptic dendritic spines were found to be retracted. Based on these results, the authors hypothesized that instead of LB-associated cell death αSyn aggregate-related synaptic dysfunction may cause neurodegeneration. Although this concept has not been examined in iLBD, it suggests that the traditional neuropathological staging (assessing somatic LBs) may not capture the true onset or progression of LBP, thus limiting its validity [95, 130, 223].

Conclusion

Here, we summarized cellular and molecular changes occurring in the SNc of iLBD (and pPD) cases. The body of previous work collectively demonstrates numerous pathological changes that appear to precede LBP in PD. These results challenge the current understanding of PD disease progression and the impact of LBP and, in a broader sense, the development of therapeutic strategies that focus on targeting αSyn [224,225,226]. Therefore, our review may provide a starting point for future studies, which will have to further examin and connect these initial molecular changes occurring in early PD. Our work will support the investigation of novel molecular targets that could halt disease progression before the known neuropathological signs begin to show.

Availability of data and materials

N.a.

Abbreviations

PD:

Parkinson’s disease

DA:

Dopaminergic

SNc:

Substantia nigra pars compacta

LBs:

Lewy bodies

αSyn:

αSynuclein (Protein)

LBP:

Lewy body pathology

CNS:

Central nervous system

iLBD:

Incidental Lewy body disease

DMV:

Dorsal motor nucleus of the vagus nerve

AD:

Alzheimer’s disease

LRRK2:

Leucine-rich repeat kinase 2

PARK2:

Parkinson juvenile disease protein 2

STED:

Stimulated emission depletion

PINK1:

PTEN-induced kinase 1

cPD:

Clinical PD

pPD:

Prodromal PD

TH:

Tyrosine Hydroxylase

SNARE:

Soluble N-ethylmaleimide-sensitive-factor attachment receptor

SNAP29:

Synaptosome Associated Protein 29

TLR-2:

Toll-like-Rezeptor 2

Cd68:

Cluster of Differentiation

ACC:

Anterior cingulate cortex

PAR2:

Protease activated receptor 2

MCM2:

Minichromosome maintenance protein 2

HC:

Hippocampus

LC:

Locus caeruleus

VMAT2:

Vesicular monoamine transporter 2

DJ-1:

Protein deglycase 1

Rab38B:

Ras-related in brain

mTOR:

Mammalian Target of Rapamycin

eIF2:

Elongation initiation factor

mtDNA:

Mitochondrial DNA

GAPDH:

Glyceraldehyde dehydrogenase

GSH:

Glutathion

FMRP:

Fragile X mental retardation protein

RBD:

REM sleep behaviour disorder

iRBD:

Idiopathic RBD

DaT:

Dopamine Transporter

PET:

Positron emission tomography

TSPO:

18 KDa translocator protein

(pre-)LBD:

(Pre-)Lewy body disease

sNfL:

Serum neurofilament light chain

DaT:

Dopamine Transporter

GBA:

Glucocerebrosidase

SNCA:

Alpha-Synculein (Gene)

References

  1. Berg D, Postuma RB, Bloem B, Chan P, Dubois B, Gasser T, et al. Time to redefine PD? Introductory statement of the MDS task force on the definition of Parkinson’s disease. Mov Disord. 2014;29:454–62.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Hornykiewicz O. Dopamine miracle: from brain homogenate to dopamine replacement. Mov Disord. 2002;17:501–8.

    Article  PubMed  Google Scholar 

  3. Dijkstra AA, Voorn P, Berendse HW, Groenewegen HJ, Bank NB, Rozemuller AJM, et al. Stage-dependent nigral neuronal loss in incidental Lewy body and Parkinson’s disease. Mov Disord. 2014;29:1244–51.

    Article  PubMed  Google Scholar 

  4. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–40.

    Article  CAS  PubMed  Google Scholar 

  5. Shahmoradian SH, Lewis AJ, Genoud C, Hench J, Moors TE, Navarro PP, et al. Lewy pathology in Parkinson’s disease consists of crowded organelles and lipid membranes. Nat Neurosci. 2019;22:1099–109.

    Article  CAS  PubMed  Google Scholar 

  6. Chartier-Harlin MC, Chartier-Harlin M-C, Kachergus J, Kachergus J, Roumier C, Roumier C, et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet. 2004;364:1167–9.

    Article  CAS  PubMed  Google Scholar 

  7. Krüger R, Kuhn W, Müller T, Woitalla D, Graeber M, Kösel S, et al. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat Genet. 1998;18:106–8.

    Article  PubMed  Google Scholar 

  8. 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 (New York, NY). 1997;276:2045–7.

    Article  CAS  Google Scholar 

  9. Papapetropoulos S, Adi N, Ellul J, Argyriou AA, Chroni E. A Prospective study of familial versus sporadic Parkinson’s disease. Neurodegener Dis. 2007;4:424–7.

    Article  PubMed  Google Scholar 

  10. Nalls MA, Pankratz N, Lill CM, Do CB, Hernandez DG, Saad M, et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat Genet. 2014;46:989–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Braak H, Tredici KD. neuropathological staging of brain pathology in sporadic Parkinson’s disease: separating the wheat from the chaff. J Park Dis. 2017;7:S73-87.

    Google Scholar 

  12. Goedert M, Spillantini MG, Tredici KD, Braak H. 100 years of Lewy pathology. 2013.

    Book  Google Scholar 

  13. Braak H, Braak H, Tredici KD, Tredici KD. Neuroanatomy and pathology of sporadic Parkinson’s disease. Adv Anat Embryol Cell Biol. 2009;201:1–119.

    PubMed  Google Scholar 

  14. Jellinger KA. Neuropathology and pathogenesis of extrapyramidal movement disorders: a critical update—I Hypokinetic-rigid movement disorders. J Neural Transm. 2019;126:933–95.

    Article  PubMed  Google Scholar 

  15. Sian-Hulsmann J, Monoranu C, Strobel S, Riederer P. Lewy bodies: a spectator or salient killer? Cns Neurol Disord Drug Targets. 2015;14:947–55.

    Article  CAS  PubMed  Google Scholar 

  16. van de Berg WDJ, Hepp DH, Dijkstra AA, Rozemuller JAM, Berendse HW, Foncke E. Patterns of alpha-synuclein pathology in incidental cases and clinical subtypes of Parkinson’s disease. Parkinsonism Relat D. 2012;18:S28-30.

    Article  Google Scholar 

  17. Braak H, Tredici KD, Rüb U, de Vos RAI, Steur ENHJ, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211.

    Article  PubMed  Google Scholar 

  18. Braak H, Ghebremedhin E, Rüb U, Bratzke H, Tredici KD. Stages in the development of Parkinson’s disease-related pathology. Cell Tissue Res. 2004;318:121–34.

    Article  PubMed  Google Scholar 

  19. 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;117:613–34.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Li J-Y, Li JY, Englund E, Englund E, Holton JL, Holton JL, et al. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nat Med. 2008;14:501–3.

    Article  CAS  PubMed  Google Scholar 

  21. Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson’s disease. Nat Med. 2008;14:504–6.

    Article  CAS  PubMed  Google Scholar 

  22. Rey NL, Steiner JA, Maroof N, Luk KC, Madaj Z, Trojanowski JQ, et al. Widespread transneuronal propagation of α-synucleinopathy triggered in olfactory bulb mimics prodromal Parkinson’s disease. J Exp Med. 2016;213:1759–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P, Trojanowski JQ, et al. Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science (New York, NY). 2012;338:949–53.

    Article  CAS  Google Scholar 

  24. Peelaerts W, Bousset L, der Perren AV, Moskalyuk A, Pulizzi R, Giugliano M, et al. α-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature. 2015;522:340–4.

    Article  CAS  PubMed  Google Scholar 

  25. Masuda-Suzukake M, Nonaka T, Hosokawa M, Oikawa T, Arai T, Akiyama H, et al. Prion-like spreading of pathological α-synuclein in brain. Brain. 2013;136:1128–38.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Recasens A, Dehay B, Bové J, Carballo-Carbajal I, Dovero S, Pérez-Villalba A, et al. Lewy body extracts from Parkinson disease brains trigger α-synuclein pathology and neurodegeneration in mice and monkeys. Ann Neurol. 2014;75:351–62.

    Article  CAS  PubMed  Google Scholar 

  27. Shimozawa A, Ono M, Takahara D, Tarutani A, Imura S, Masuda-Suzukake M, et al. Propagation of pathological α-synuclein in marmoset brain. Acta Neuropathol Commun. 2017;5:12.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Gibb WR, Lees AJ. The relevance of the Lewy body to the pathogenesis of idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1988;51:745–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jellinger KA. A critical reappraisal of current staging of Lewy-related pathology in human brain. Acta Neuropathol. 2008;116:1–16.

    Article  CAS  PubMed  Google Scholar 

  30. Parkkinen L, Pirttilä T, Alafuzoff I. Applicability of current staging/categorization of α-synuclein pathology and their clinical relevance. Acta Neuropathol. 2008;115:399–407.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Ansorge O, Daniel SE, Pearce RK. Neuronal loss and plasticity in the supraoptic nucleus in Parkinson’s disease. Neurology. 1997;49:610–3.

    Article  CAS  PubMed  Google Scholar 

  32. MacDonald V, Halliday GM. Selective loss of pyramidal neurons in the pre-supplementary motor cortex in Parkinson’s disease. Mov Disord. 2002;17:1166–73.

    Article  PubMed  Google Scholar 

  33. Pedersen KM, Marner L, Pakkenberg H, Pakkenberg B. No global loss of neocortical neurons in Parkinson’s disease: a quantitative stereological study. Mov Disord. 2005;20:164–71.

    Article  PubMed  Google Scholar 

  34. Halliday GM, Li YW, Blumbergs PC, Joh TH, Cotton RG, Howe PR, et al. Neuropathology of immunohistochemically identified brainstem neurons in Parkinson’s disease. Ann Neurol. 1990;27:373–85.

    Article  CAS  PubMed  Google Scholar 

  35. Thannickal TC, Lai Y-Y, Siegel JM. Hypocretin (orexin) cell loss in Parkinson’s disease. Brain. 2007;130:1586–95.

    Article  PubMed  Google Scholar 

  36. Kremer HP, Bots GT. Lewy bodies in the lateral hypothalamus: do they imply neuronal loss? Mov Disord. 1993;8:315–20.

    Article  CAS  PubMed  Google Scholar 

  37. Iacono D, Geraci-Erck M, Rabin ML, Adler CH, Serrano G, Beach TG, et al. Parkinson disease and incidental Lewy body disease: just a question of time? Neurology. 2015;85:1670–9.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Schneider SA, Alcalay RN. Neuropathology of genetic synucleinopathies with parkinsonism: review of the literature. Mov Disord. 2017;32:1504–23.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Kalia LV, Lang AE, Hazrati L-N, Fujioka S, Wszolek ZK, Dickson DW, et al. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease. JAMA Neurol. 2015;72:100–5.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Doherty KM, Silveira-Moriyama L, Parkkinen L, Healy DG, Farrell M, Mencacci NE, et al. Parkin disease: a clinicopathologic entity? JAMA Neurol. 2013;70:571–9.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Wakabayashi K, Tanji K, Mori F, Takahashi H. The Lewy body in Parkinson’s disease: molecules implicated in the formation and degradation of α-synuclein aggregates: molecular components of Lewy body. Neuropathology. 2007;27:494–506.

    Article  PubMed  Google Scholar 

  42. Tompkins MM, Hill WD. Contribution of somal Lewy bodies to neuronal death. Brain Res. 1997;775:24–9.

    Article  CAS  PubMed  Google Scholar 

  43. Bergeron C, Petrunka C, Weyer L, Pollanen MS. Altered neurofilament expression does not contribute to Lewy body formation. Am J Pathology. 1996;148:267–72.

    CAS  Google Scholar 

  44. Hill WD. Altered neurofilament expression does not contribute to Lewy body formation. Am J Pathol. 1996;149:728–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Javoy-Agid F, Hirsch EC, Dumas S, Duyckaerts C, Mallet J, Agid Y. Decreased tyrosine hydroxylase messenger RNA in the surviving dopamine neurons of the substantia nigra in parkinson’s disease: An in situ hybridization study. Neuroscience. 1990;38:245–53.

    Article  CAS  PubMed  Google Scholar 

  46. Patt S, Gertz HJ, Gerhard L, Cervós-Navarro J. Pathological changes in dendrites of substantia nigra neurons in Parkinson’s disease: a Golgi study. Histol Histopathol. 1991;6:373–80.

    CAS  PubMed  Google Scholar 

  47. Gomez-Isla T, Growdon WB, McNamara M, Newell K, Gomez-Tortosa E, Hedley-Whyte ET, et al. Clinicopathologic correlates in temporal cortex in dementia with Lewy bodies. Neurology. 1999;53:2003–2003.

    Article  CAS  PubMed  Google Scholar 

  48. Gomez-Tortosa E, Newell K, Irizarry MC, Albert M, Growdon JH, Hyman BT. Clinical and quantitative pathologic correlates of dementia with Lewy bodies. Neurology. 1999;53:1284–1284.

    Article  CAS  PubMed  Google Scholar 

  49. Mattila PM, Rinne JO, Helenius H, Dickson DW, Röyttä M, Mattila PM. Alpha-synuclein-immunoreactive cortical Lewy bodies are associated with cognitive impairment in Parkinson’s disease. Acta Neuropathol. 2000;100:285–90.

    Article  CAS  PubMed  Google Scholar 

  50. Weisman D, Cho M, Taylor C, Adame A, Thal LJ, Hansen LA. In dementia with Lewy bodies, Braak stage determines phenotype, not Lewy body distribution. Neurology. 2007;69:356–9.

    Article  CAS  PubMed  Google Scholar 

  51. Dong X, Mondello S, Kobeissy F, Talih F, Ferri R, Mechref Y. LC–MS/MS glycomics of idiopathic rapid eye movement sleep behavior disorder. Electrophoresis. 2018;39:3096–103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Mondello S, Kobeissy F, Mechref Y, Zhao J, Talih FR, Cosentino F, et al. Novel biomarker signatures for idiopathic REM sleep behavior disorder: a proteomic and system biology approach. Neurology. 2018;91:e1710–5.

    Article  PubMed  Google Scholar 

  53. Schmidt ML, Martin JA, Lee VM-Y, Trojanowski JQ. Convergence of Lewy bodies and neurofibrillary tangles in amygdala neurons of Alzheimer’s disease and Lewy body disorders. Acta Neuropathol. 1996;91:475–81.

    Article  CAS  PubMed  Google Scholar 

  54. Schulz-Schaeffer WJ. Neurodegeneration in Parkinson disease: Moving Lewy bodies out of focus. Neurology. 2012;79:2298–9.

    Article  PubMed  Google Scholar 

  55. Olanow CW, Perl DP, DeMartino GN, McNaught KSP. Lewy-body formation is an aggresome-related process: a hypothesis. Lancet Neurol. 2004;3:496–503.

    Article  PubMed  Google Scholar 

  56. Lashuel HA. Do Lewy bodies contain alpha-synuclein fibrils? and Does it matter? A brief history and critical analysis of recent reports. Neurobiol Dis. 2020;141:104876.

    Article  CAS  PubMed  Google Scholar 

  57. Valente EM, Abou-Sleiman PM, Caputo V, Muqit MMK, Harvey K, Gispert S, et al. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science. 2004;304:1158–60.

    Article  CAS  PubMed  Google Scholar 

  58. Sidransky E, Nalls MA, Aasly JO, Aharon-Peretz J, Annesi G, Barbosa ER, et al. Multicenter analysis of glucocerebrosidase mutations in Parkinson’s disease. New Engl J Medicine. 2009;361:1651–61.

    Article  CAS  Google Scholar 

  59. Robak LA, Jansen IE, van Rooij J, Uitterlinden AG, Kraaij R, Jankovic J, et al. Excessive burden of lysosomal storage disorder gene variants in Parkinson’s disease. Brain. 2017;140:3191–203.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Rocha EM, Smith GA, Park E, Cao H, Brown E, Hallett P, et al. Progressive decline of glucocerebrosidase in aging and Parkinson’s disease. Ann Clin Transl Neur. 2015;2:433–8.

    Article  Google Scholar 

  61. Rocha EM, Smith GA, Park E, Cao H, Graham A-R, Brown E, et al. Sustained systemic glucocerebrosidase inhibition induces brain α-synuclein aggregation, microglia and complement C1q activation in mice. Antioxid Redox Sign. 2015;23:550–64.

    Article  CAS  Google Scholar 

  62. Tan MMX, Malek N, Lawton MA, Hubbard L, Pittman AM, Joseph T, et al. Genetic analysis of Mendelian mutations in a large UK population-based Parkinson’s disease study. Brain. 2019;142:2828–44.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Jellinger KA. Is Braak staging valid for all types of Parkinson’s disease? J Neural Transm Vienna Austria. 1996;2018(126):423–31.

    Google Scholar 

  64. Saito Y, Ruberu NN, Sawabe M, Arai T, Kazama H, Hosoi T, et al. Lewy body-related α-synucleinopathy in aging. J Neuropathol Exp Neurology. 2004;63:742–9.

    Article  Google Scholar 

  65. Bloch A, Probst A, Bissig H, Adams H, Tolnay M. Alpha-synuclein pathology of the spinal and peripheral autonomic nervous system in neurologically unimpaired elderly subjects. Neuropathol Appl Neurobiol. 2006;32:284–95.

    Article  CAS  PubMed  Google Scholar 

  66. Caviness JN. Presymptomatic Parkinson’s disease: the Arizona experience. Parkinsonism Relat D. 2012;18:S203–6.

    Article  Google Scholar 

  67. Adler CH, Connor DJ, Hentz JG, Sabbagh MN, Caviness JN, Shill HA, et al. Incidental Lewy body disease: clinical comparison to a control cohort. Movement Disord. 2010;25:642–6.

    Article  PubMed  Google Scholar 

  68. Driver-Dunckley E, Adler CH, Hentz JG, Dugger BN, Shill HA, Caviness JN, et al. Olfactory dysfunction in incidental Lewy body disease and Parkinson’s disease. Parkinsonism Relat D. 2014;20:1260–2.

    Article  Google Scholar 

  69. Adler CH, Dugger BN, Hentz JG, Hinni ML, Lott DG, Driver-Dunckley E, et al. Peripheral synucleinopathy in early Parkinson’s disease: Submandibular gland needle biopsy findings. Mov Disord. 2017;32(5):722-723. https://doi.org/10.1002/mds.27044. PMID: 28513078.

  70. Jellinger KA. Lewy body-related ?-synucleinopathy in the aged human brain. J Neural Transm. 2004;111:1219–35.

    Article  CAS  PubMed  Google Scholar 

  71. Dickson DW, Fujishiro H, DelleDonne A, Menke J, Ahmed Z, Klos KJ, et al. Evidence that incidental Lewy body disease is pre-symptomatic Parkinson’s disease. Acta Neuropathol. 2008;115:437–44.

    Article  PubMed  Google Scholar 

  72. Greffard S, Verny M, Bonnet A-M, Beinis J-Y, Gallinari C, Meaume S, et al. Motor score of the unified parkinson disease rating scale as a good predictor of Lewy body-associated neuronal loss in the substantia Nigra. Arch Neurol Chicago. 2006;63:584–8.

    Article  PubMed  Google Scholar 

  73. Kordower JH, Olanow CW, Dodiya HB, Chu Y, Beach TG, Adler CH, et al. Disease duration and the integrity of the nigrostriatal system in Parkinson’s disease. Brain. 2013;136:2419–31.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Chu Y, Buchman AS, Olanow CW, Kordower JH. Do subjects with minimal motor features have prodromal Parkinson disease? Ann Neurol. 2018;83:562–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Fearnley JM, Lees AJ. Ageing, and Parkinson’s disease: substantia nigra regional selectivity. Brain. 1991;114:2283–301.

    Article  PubMed  Google Scholar 

  76. Damier P, Hirsch EC, Agid Y, Graybiel AM. The substantia nigra of the human brain. II. Patterns of loss of dopamine-containing neurons in Parkinson’s disease. Brain. 1999;122(Pt 8):1437–48.

    Article  PubMed  Google Scholar 

  77. Halliday GM, Song YJC, Harding AJ. Striatal β-amyloid in dementia with Lewy bodies but not Parkinson’s disease. J Neural Trans (Vienna, Austria: 1996). 2011;118:713–9.

    Article  CAS  Google Scholar 

  78. DelleDonne A, Klos KJ, Fujishiro H, Ahmed Z, Parisi JE, Josephs KA, et al. Incidental Lewy body disease and preclinical Parkinson disease. Arch Neurol. 2008;65:1074–80.

    Article  PubMed  Google Scholar 

  79. Milber JM, Noorigian JV, Morley JF, Petrovitch H, White L, Ross GW, et al. Lewy pathology is not the first sign of degeneration in vulnerable neurons in Parkinson disease. Neurology. 2012;79:2307–14.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Jellinger KA. A critical evaluation of current staging of α-synuclein pathology in Lewy body disorders. BBA Mol Basis Dis. 2009;1792:730–40.

    Article  CAS  Google Scholar 

  81. Iranzo A, Gelpi E, Tolosa E, Molinuevo JL, Serradell M, Gaig C, et al. Neuropathology of prodromal Lewy body disease. Mov Disord. 2014;29:410–5.

    Article  PubMed  Google Scholar 

  82. Beach TG, Adler CH, Sue LI, Peirce JB, Bachalakuri J, Dalsing-Hernandez JE, et al. Reduced striatal tyrosine hydroxylase in incidental Lewy body disease. Acta Neuropathol. 2008;115:445–51.

    Article  CAS  PubMed  Google Scholar 

  83. Dalfó E, Ferrer I. Early α-synuclein lipoxidation in neocortex in Lewy body diseases. Neurobiol Aging. 2008;29:408–17.

    Article  PubMed  Google Scholar 

  84. Ferrer I, Martinez A, Blanco R, Dalfó E, Carmona M. Neuropathology of sporadic Parkinson disease before the appearance of parkinsonism: preclinical Parkinson disease. J Neural Transm (Vienna, Austria: 1996). 2011;118:821–39.

    Article  Google Scholar 

  85. Koziorowski D, Friedman A, Arosio P, Santambrogio P, Dziewulska D. ELISA reveals a difference in the structure of substantia nigra ferritin in Parkinson’s disease and incidental Lewy body compared to control. Parkinsonism Relat D. 2007;13:214–8.

    Article  Google Scholar 

  86. Tang Q, Gao P, Arzberger T, Höllerhage M, Herms J, Höglinger G, et al. Alpha-Synuclein defects autophagy by impairing SNAP29-mediated autophagosome-lysosome fusion. Cell Death Dis. 2021;12(10):854.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kuusisto E, Parkkinen L, Alafuzoff I. Morphogenesis of Lewy Bodies: Dissimilar Incorporation of α-Synuclein, Ubiquitin, and p62. J Neuropathol Exp Neurol. 2003;62:1241–53.

    Article  CAS  PubMed  Google Scholar 

  88. Doorn KJ, Moors T, Drukarch B, van de Berg WD, Lucassen PJ, van Dam A-M. Microglial phenotypes and toll-like receptor 2 in the substantia nigra and hippocampus of incidental Lewy body disease cases and Parkinson’s disease patients. Acta Neuropathol Commun. 2014;2:90.

    PubMed  PubMed Central  Google Scholar 

  89. Bradaric BD, Patel A, Schneider JA, Carvey PM, Hendey B. Evidence for angiogenesis in Parkinson’s disease, incidental Lewy body disease, and progressive supranuclear palsy. J Neural Transm. 2012;119:59–71.

    Article  Google Scholar 

  90. Hurley MJ, Durrenberger PF, Gentleman SM, Walls AF, Dexter DT. Altered expression of brain proteinase-activated receptor-2, trypsin-2 and serpin proteinase inhibitors in Parkinson’s disease. J Mol Neurosci. 2015;57:48–62.

    Article  CAS  PubMed  Google Scholar 

  91. Walker DG, Lue L-F, Serrano G, Adler CH, Caviness JN, Sue LI, et al. Altered expression patterns of inflammation-associated and trophic molecules in substantia nigra and striatum brain samples from Parkinson’s disease, incidental Lewy body disease and normal control cases. Front Neurosci Switz. 2016;9:507.

    Google Scholar 

  92. Galiano-Landeira J, Torra A, Vila M, Bové J. CD8 T cell nigral infiltration precedes synucleinopathy in early stages of Parkinson’s disease. Brain. 2020;143:3717–33.

    Article  PubMed  Google Scholar 

  93. Scherman D, Desnos C, Darchen F, Pollak P, Javoy-Agid F, Agid Y. Striatal dopamine deficiency in parkinson’s disease: role of aging. Ann Neurol. 1989;26:551–7.

    Article  CAS  PubMed  Google Scholar 

  94. Ma SY, Röyttä M, Rinne JO, Collan Y, Rinne UK. Correlation between neuromorphometry in the substantia nigra and clinical features in Parkinson’s disease using disector counts. J Neurol Sci. 1997;151:83–7.

    Article  CAS  PubMed  Google Scholar 

  95. Schulz-Schaeffer WJ. The synaptic pathology of α-synuclein aggregation in dementia with Lewy bodies, Parkinson’s disease and Parkinson’s disease dementia. Acta Neuropathol. 2010;120:131–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Dijkstra AA, Ingrassia A, de Menezes RX, van Kesteren RE, Rozemuller AJM, Heutink P, et al. Evidence for immune response, axonal dysfunction and reduced endocytosis in the substantia nigra in early stage Parkinson’s disease. PLoS One. 2015;10:e0128651.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Lin MT, Cantuti-Castelvetri I, Zheng K, Jackson KE, Tan YB, Arzberger T, et al. Somatic mitochondrial DNA mutations in early parkinson and incidental lewy body disease. Ann Neurol. 2012;71:850–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Bryois J, Skene NG, Hansen TF, Kogelman LJA, Watson HJ, Liu Z, et al. Genetic identification of cell types underlying brain complex traits yields insights into the etiology of Parkinson’s disease. Nat Genet. 2020;52:482–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Feleke R, Reynolds RH, Smith AM, Tilley B, Taliun SAG, Hardy J, et al. Cross-platform transcriptional profiling identifies common and distinct molecular pathologies in Lewy body diseases. Acta Neuropathol. 2021;142:449–74.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Santpere G, Garcia-Esparcia P, Andres-Benito P, Lorente-Galdos B, Navarro A, Ferrer I. Transcriptional network analysis in frontal cortex in Lewy body diseases with focus on dementia with Lewy bodies: transcriptional network analysis. Brain Pathol. 2017;28:315–33.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Wilhelmus MMM, Bol JGJM, Haastert ESV, Rozemuller AJM, Bu G, Drukarch B, et al. Apolipoprotein E and LRP1 increase early in Parkinson’s disease pathogenesis. Am J Pathol. 2011;179:2152–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Fernandez A, de Ceballos ML, Rose S, Jenner P, Marsden CD. Alterations in peptide levels in Parkinson’s disease and incidental Lewy body disease. Brain. 1996;119:823–30.

    Article  PubMed  Google Scholar 

  103. Jenner P, Dexter DT, Sian J, Schapira AHV, Marsden CD, Group TRKAQPDR. Oxidative stress as a cause of nigral cell death in Parkinson’s disease and incidental lewy body disease. Ann Neurol. 1992;32:S82-7.

    Article  CAS  PubMed  Google Scholar 

  104. Wilkinson H, Thomsson KA, Rebelo AL, Hilliard M, Pandit A, Rudd PM, et al. The O-Glycome of human nigrostriatal tissue and its alteration in Parkinson’s disease. J Proteome Res. 2021;20:3913–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Pearce RKB, Owen A, Daniel S, Jenner P, Marsden CD. Alterations in the distribution of glutathione in the substantia nigra in Parkinson’s disease. J Neural Transm. 1997;104:661–77.

    Article  CAS  PubMed  Google Scholar 

  106. Jenner P, Olanow CW. Oxidative stress and the pathogenesis of Parkinson’s disease. Neurology. 1996;47:161S-170S.

    Article  Google Scholar 

  107. Tan Y, Sgobio C, Arzberger T, Machleid F, Tang Q, Findeis E, et al. Loss of fragile X mental retardation protein precedes Lewy pathology in Parkinson’s disease. Acta Neuropathol. 2019;139:319–45.

    Article  PubMed  Google Scholar 

  108. Beach TG, Adler CH, Sue LI, Shill HA, Driver-Dunckley E, Mehta SH, et al. Vagus nerve and stomach synucleinopathy in Parkinson’s disease, incidental lewy body disease, and normal elderly subjects: evidence against the “Body-First” hypothesis. J Park Dis. 2021:1–11.

  109. Abbott RD, Ross GW, Petrovitch H, Tanner CM, Davis DG, Masaki KH, et al. Bowel movement frequency in late-life and incidental Lewy bodies. Movement Disord. 2007;22:1581–6.

    Article  PubMed  Google Scholar 

  110. Tredici KD, Hawkes CH, Ghebremedhin E, Braak H. Lewy pathology in the submandibular gland of individuals with incidental Lewy body disease and sporadic Parkinson’s disease. Acta Neuropathol. 2010;119:703–13.

    Article  PubMed  Google Scholar 

  111. Ghebremedhin E, Tredici KD, Langston JW, Braak H. Diminished tyrosine hydroxylase immunoreactivity in the cardiac conduction system and myocardium in Parkinson’s disease: an anatomical study. Acta Neuropathol. 2009;118:777.

    Article  CAS  PubMed  Google Scholar 

  112. Labandeira-Garcia JL, Valenzuela R, Costa-Besada MA, Villar-Cheda B, Rodriguez-Perez AI. The intracellular renin-angiotensin system: friend or foe. Some light from the dopaminergic neurons. Prog Neurobiol. 2021;199:101919.

    Article  CAS  PubMed  Google Scholar 

  113. Oro C, Qian H, Thomas WG. Type 1 angiotensin receptor pharmacology: signaling beyond G proteins. Pharmacol Therapeut. 2007;113:210–26.

    Article  CAS  Google Scholar 

  114. Lin H-C, Tseng Y-F, Shen A-L, Chao JC-J, Hsu C-Y, Lin H-L. Association of angiotensin receptor blockers with Incident Parkinson disease in patients with hypertension: a retrospective cohort study. Am J Medicine. 2022;135:1001–7.

    Article  CAS  Google Scholar 

  115. Jo Y, Kim S, Ye BS, Lee E, Yu YM. Protective Effect of Renin-Angiotensin system inhibitors on Parkinson’s disease: a nationwide cohort study. Front Pharmacol. 2022;13:837890.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Forloni G. Alpha Synuclein: Neurodegeneration and Inflammation. Int J Mol Sci. 2023;24:5914.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Danzer KM, Kranich LR, Ruf WP, Cagsal-Getkin O, Winslow AR, Zhu L, et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol Neurodegener. 2012;7:42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Sekiya H, Tsuji A, Hashimoto Y, Takata M, Koga S, Nishida K, et al. Discrepancy between distribution of alpha-synuclein oligomers and Lewy-related pathology in Parkinson’s disease. Acta Neuropathol Commun. 2022;10:133.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Kumar ST, Jagannath S, Francois C, Vanderstichele H, Stoops E, Lashuel HA. How specific are the conformation-specific α-synuclein antibodies? Characterization and validation of 16 α-synuclein conformation-specific antibodies using well-characterized preparations of α-synuclein monomers, fibrils and oligomers with distinct structures and morphology. Neurobiol Dis. 2020;146:105086.

    Article  CAS  PubMed  Google Scholar 

  120. Stokholm MG, Iranzo A, Østergaard K, Serradell M, Otto M, Svendsen KB, et al. Assessment of neuroinflammation in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a case-control study. Lancet Neurol. 2017;16:789–96.

    Article  PubMed  Google Scholar 

  121. Duffy MF, Collier TJ, Patterson JR, Kemp CJ, Luk KC, Tansey MG, et al. Lewy body-like alpha-synuclein inclusions trigger reactive microgliosis prior to nigral degeneration. J Neuroinflamm. 2018;15:129.

    Article  Google Scholar 

  122. Carroll CB, Webb D, Stevens KN, Vickery J, Eyre V, Ball S, et al. Simvastatin as a neuroprotective treatment for Parkinson’s disease (PD STAT): protocol for a double-blind, randomised, placebo-controlled futility study. BMJ Open. 2019;9:e029740.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Selley ML. Simvastatin prevents 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced striatal dopamine depletion and protein tyrosine nitration in mice. Brain Res. 2005;1037:1–6.

    Article  CAS  PubMed  Google Scholar 

  124. Ghosh A, Roy A, Matras J, Brahmachari S, Gendelman HE, Pahan K. Simvastatin inhibits the activation of p21ras and prevents the loss of dopaminergic neurons in a mouse model of Parkinson’s disease. J Neurosci. 2009;29:13543–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. The PD-STAT/Simvastatin study results. Available from: https://cureparkinsons.org.uk/2020/09/post-alternative-layout-2-without-image-2/.

  126. Greenland JC, Cutting E, Kadyan S, Bond S, Chhabra A, Williams-Gray CH. Azathioprine immunosuppression and disease modification in Parkinson’s disease (AZA-PD): a randomised double-blind placebo-controlled phase II trial protocol. BMJ Open. 2020;10:e040527.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Su Q, Ng WL, Goh SY, Gulam MY, Wang L-F, Tan E-K, et al. Targeting the inflammasome in Parkinson’s disease. Front Aging Neurosci. 2022;14: 957705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Overk CR, Masliah E. Pathogenesis of synaptic degeneration in Alzheimer’s disease and Lewy body disease. Biochem Pharmacol. 2014;88:508–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Miller GW, Erickson JD, Perez JT, Penland SN, Mash DC, Rye DB, et al. Immunochemical Analysis of Vesicular Monoamine Transporter (VMAT2) Protein in Parkinson’s Disease. Exp Neurol. 1999;156:138–48.

    Article  CAS  PubMed  Google Scholar 

  130. Kramer ML, Schulz-Schaeffer WJ. Presynaptic -synuclein aggregates, not Lewy bodies, cause neurodegeneration in dementia with Lewy bodies. J Neurosci. 2007;27:1405–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Burke RE, O’Malley K. Axon degeneration in Parkinson’s disease. Exp Neurol. 2012;246:72–83.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Cheng H-C, Ulane CM, Burke RE. Clinical progression in Parkinson disease and the neurobiology of axons. Ann Neurol. 2010;67:715–25.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Soukup S-F, Vanhauwaert R, Verstreken P. Parkinson’s disease: convergence on synaptic homeostasis. Embo J. 2018;37(18):e98960.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Nguyen M, Wong YC, Ysselstein D, Severino A, Krainc D. Synaptic, mitochondrial, and lysosomal dysfunction in Parkinson’s disease. Trends Neurosci. 2018;42:140–9.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Bellucci A, Zaltieri M, Navarria L, Grigoletto J, Missale C, Spano P. From α-synuclein to synaptic dysfunctions: New insights into the pathophysiology of Parkinson’s disease. Brain Res. 2012;1476:183–202.

    Article  CAS  PubMed  Google Scholar 

  136. Nikolaus S, Antke C, Müller H-W. In vivo imaging of synaptic function in the central nervous system. Behav Brain Res. 2009;204:1–31.

    Article  PubMed  Google Scholar 

  137. Revuelta GJ, Rosso A, Lippa CF. Neuritic Pathology as a Correlate of Synaptic Loss in Dementia With Lewy Bodies. Am J Alzheimer’s Dis Other Dementiasr. 2008;23:97–102.

    Article  Google Scholar 

  138. McNeill TH, Brown SA, Rafols JA, Shoulson I. Atrophy of medium spiny I striatal dendrites in advanced Parkinson’s disease. Brain Res. 1988;455:148–52.

    Article  CAS  PubMed  Google Scholar 

  139. Zaja-Milatovic S, Milatovic D, Schantz AM, Zhang J, Montine KS, Samii A, et al. Dendritic degeneration in neostriatal medium spiny neurons in Parkinson disease. Neurology. 2005;64:545–7.

    Article  CAS  PubMed  Google Scholar 

  140. Kitada T, Tong Y, Gautier CA, Shen J. Absence of nigral degeneration in aged parkin/DJ-1/PINK1 triple knockout mice. J Neurochem. 2009;111:696–702.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Scherfler C, Khan NL, Pavese N, Eunson L, Graham E, Lees AJ, et al. Striatal and cortical pre- and postsynaptic dopaminergic dysfunction in sporadic parkin-linked parkinsonism. Brain. 2004;127:1332–42.

    Article  PubMed  Google Scholar 

  142. Chou J-S, Chen C-Y, Chen Y-L, Weng Y-H, Yeh T-H, Lu C-S, et al. (G2019S) LRRK2 causes early-phase dysfunction of SNpc dopaminergic neurons and impairment of corticostriatal long-term depression in the PD transgenic mouse. Neurobiol Dis. 2014;68:190–9.

    Article  CAS  PubMed  Google Scholar 

  143. Beccano-Kelly DA, Volta M, Munsie LN, Paschall SA, Tatarnikov I, Co K, et al. LRRK2 overexpression alters glutamatergic presynaptic plasticity, striatal dopamine tone, postsynaptic signal transduction, motor activity and memory. Hum Mol Genet. 2014;24:1336–49.

    Article  PubMed  Google Scholar 

  144. Madeo G, Schirinzi T, Martella G, Latagliata EC, Puglisi F, Shen J, et al. PINK1 heterozygous mutations induce subtle alterations in dopamine-dependent synaptic plasticity: synaptic plasticity in Pink1 +/− Mice. Movement Disord. 2013;29:41–53.

    Article  PubMed  Google Scholar 

  145. Wilson GR, Sim JCH, McLean C, Giannandrea M, Galea CA, Riseley JR, et al. Mutations in RAB39B cause X-linked intellectual disability and early-onset Parkinson disease with α-synuclein pathology. Am J Hum Genetics. 2014;95:729–35.

    Article  CAS  Google Scholar 

  146. Linazasoro G. Classical Parkinson disease versus Parkinson complex ? Reflections against staging and in favour of heterogeneity. Eur J Neurol. 2007;14:721–8.

    Article  CAS  PubMed  Google Scholar 

  147. Hunn BHM, Cragg SJ, Bolam JP, Spillantini M-G, Wade-Martins R. Impaired intracellular trafficking defines early Parkinson’s disease. Trends Neurosci. 2015;38:178–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Lesnick TG, Papapetropoulos S, Mash DC, Ffrench-Mullen J, Shehadeh L, de Andrade M, et al. A Genomic pathway approach to a complex disease: axon guidance and Parkinson disease. Plos Genet. 2007;3:e98.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Moran LB, Duke DC, Deprez M, Dexter DT, Pearce RKB, Graeber MB. Whole genome expression profiling of the medial and lateral substantia nigra in Parkinson’s disease. Neurogenetics. 2006;7:1–11.

    Article  CAS  PubMed  Google Scholar 

  150. Gómez A, Ferrer I. Increased oxidation of certain glycolysis and energy metabolism enzymes in the frontal cortex in Lewy body diseases. J Neurosci Res. 2009;87:1002–13.

    Article  PubMed  Google Scholar 

  151. Contractor A, Klyachko VA, Portera-Cailliau C. Altered neuronal and circuit excitability in fragile X syndrome. Neuron. 2015;87:699–715.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Richter JD, Zhao X. The molecular biology of FMRP: new insights into fragile X syndrome. Nat Rev Neurosci. 2021;22:209–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Tredici KD, Duda JE. Peripheral Lewy body pathology in Parkinson’s disease and incidental Lewy body disease: four cases. J Neurol Sci. 2011;310:100–6.

    Article  PubMed  Google Scholar 

  154. Iwanaga K, Wakabayashi K, Yoshimoto M, Tomita I, Satoh H, Takashima H, et al. Lewy body-type degeneration in cardiac plexus in Parkinson’s and incidental Lewy body diseases. Neurology. 1999;52:1269–1269.

    Article  CAS  PubMed  Google Scholar 

  155. Tamura T, Yoshida M, Hashizume Y, Sobue G. Lewy body-related α-synucleinopathy in the spinal cord of cases with incidental Lewy body disease. Neuropathology. 2012;32:13–22.

    Article  PubMed  Google Scholar 

  156. Berg D, Postuma RB, Adler CH, Bloem BR, Chan P, Dubois B, et al. MDS research criteria for prodromal Parkinson’s disease: MDS Criteria for Prodromal PD. Movement Disord. 2015;30:1600–11.

    Article  PubMed  Google Scholar 

  157. Berg D, Borghammer P, Fereshtehnejad S-M, Heinzel S, Horsager J, Schaeffer E, et al. Prodromal Parkinson disease subtypes — key to understanding heterogeneity. Nat Rev Neurol. 2021;17(6):349-361. https://doi.org/10.1038/s41582-021-00486-9. Epub 2021 Apr 20. PMID: 33879872.

  158. Fereshtehnejad S-M, Yao C, Pelletier A, Montplaisir JY, Gagnon J-F, Postuma RB. Evolution of prodromal Parkinson’s disease and dementia with Lewy bodies: a prospective study. Brain. 2019;142:2051–67.

    Article  PubMed  Google Scholar 

  159. Schrag A, Horsfall L, Walters K, Noyce A, Petersen I. Prediagnostic presentations of Parkinson’s disease in primary care: a case-control study. Lancet Neurol. 2015;14:57–64.

    Article  PubMed  Google Scholar 

  160. Rahayel S, Postuma RB, Montplaisir J, Bedetti C, Brambati S, Carrier J, et al. Abnormal gray matter shape, thickness, and volume in the motor cortico-subcortical loop in idiopathic rapid eye movement sleep behavior disorder: association with clinical and motor features. Cereb Cortex. 2017;28:658–71.

    Article  Google Scholar 

  161. Barber TR, Griffanti L, Bradley KM, McGowan DR, Lo C, Mackay CE, et al. Nigrosome 1 imaging in REM sleep behavior disorder and its association with dopaminergic decline. Ann Clin Transl Neur. 2020;7:26–35.

    Article  CAS  Google Scholar 

  162. Iranzo A, Fairfoul G, Ayudhaya ACN, Serradell M, Gelpi E, Vilaseca I, et al. Detection of α-synuclein in CSF by RT-QuIC in patients with isolated rapid-eye-movement sleep behaviour disorder: a longitudinal observational study. Lancet Neurol. 2021;20:203–12.

    Article  CAS  PubMed  Google Scholar 

  163. Galbiati A, Verga L, Giora E, Zucconi M, Ferini-Strambi L. The risk of neurodegeneration in REM sleep behavior disorder: a systematic review and meta-analysis of longitudinal studies. Sleep Med Rev. 2019;43:37–46.

    Article  PubMed  Google Scholar 

  164. Dauvilliers Y, Schenck CH, Postuma RB, Iranzo A, Luppi P-H, Plazzi G, et al. REM sleep behaviour disorder. Nat Rev Dis Primers. 2018;4:19.

    Article  PubMed  Google Scholar 

  165. Chen T-Z, Xu G-J, Zhou G-A, Wang J-R, Chan P, Du Y-F. Postural sway in idiopathic rapid eye movement sleep behavior disorder: a potential marker of prodromal Parkinson′s disease. Brain Res. 2014;1559:26–32.

    Article  CAS  PubMed  Google Scholar 

  166. Alibiglou L, Videnovic A, Planetta PJ, Vaillancourt DE, MacKinnon CD. Subliminal gait initiation deficits in rapid eye movement sleep behavior disorder: a harbinger of freezing of gait? Mov Disord. 2016;31:1711–9.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Postuma RB, Gagnon JF, Vendette M, Montplaisir JY. Markers of neurodegeneration in idiopathic rapid eye movement sleep behaviour disorder and Parkinson’s disease. Brain. 2009;132:3298–307.

    Article  CAS  PubMed  Google Scholar 

  168. Postuma RB, Lang AE, Gagnon JF, Pelletier A, Montplaisir JY. How does parkinsonism start? Prodromal parkinsonism motor changes in idiopathic REM sleep behaviour disorder. Brain. 2012;135:1860–70.

    Article  CAS  PubMed  Google Scholar 

  169. Eisensehr I, Linke R, Noachtar S, Schwarz J, Gildehaus FJ, Tatsch K. Reduced striatal dopamine transporters in idiopathic rapid eye movement sleep behaviour disorderComparison with Parkinson’s disease and controls. Brain. 2000;123:1155–60.

    Article  PubMed  Google Scholar 

  170. Bauckneht M, Chincarini A, Carli FD, Terzaghi M, Morbelli S, Nobili F, et al. Presynaptic dopaminergic neuroimaging in REM sleep behavior disorder: a systematic review and meta-analysis. Sleep Med Rev. 2018;41:266–74.

    Article  PubMed  Google Scholar 

  171. Beauchamp LC, Villemagne VL, Finkelstein DI, Doré V, Bush AI, Barnham KJ, et al. Reduced striatal vesicular monoamine transporter 2 in REM sleep behavior disorder: imaging prodromal parkinsonism. Sci Rep-uk. 2020;10:17631.

    Article  CAS  Google Scholar 

  172. Huang Z, Jiang C, Li L, Xu Q, Ge J, Li M, et al. Correlations between dopaminergic dysfunction and abnormal metabolic network activity in REM sleep behavior disorder. J Cereb Blood Flow Metab. 2019;40:552–62.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Marzi RD, Seppi K, Högl B, Müller C, Scherfler C, Stefani A, et al. Loss of dorsolateral nigral hyperintensity on 3.0 tesla susceptibility-weighted imaging in idiopathic rapid eye movement sleep behavior disorder. Ann Neurol. 2016;79:1026–30.

    Article  PubMed  Google Scholar 

  174. Rolinski M, Griffanti L, Piccini P, Roussakis AA, Szewczyk-Krolikowski K, Menke RA, et al. Basal ganglia dysfunction in idiopathic REM sleep behaviour disorder parallels that in early Parkinson’s disease. Brain. 2016;139:2224–34.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Ellmore TM, Castriotta RJ, Hendley KL, Aalbers BM, Furr-Stimming E, Hood AJ, et al. Altered nigrostriatal and nigrocortical functional connectivity in rapid eye movement sleep behavior disorder. Sleep. 2013;36:1885–92.

    Article  PubMed  PubMed Central  Google Scholar 

  176. Ehrminger M, Latimier A, Pyatigorskaya N, Garcia-Lorenzo D, Leu-Semenescu S, Vidailhet M, et al. The coeruleus/subcoeruleus complex in idiopathic rapid eye movement sleep behaviour disorder. Brain. 2016;139:1180–8.

    Article  PubMed  Google Scholar 

  177. Iranzo A, Lomeña F, Stockner H, Valldeoriola F, Vilaseca I, Salamero M, et al. Decreased striatal dopamine transporter uptake and substantia nigra hyperechogenicity as risk markers of synucleinopathy in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a prospective study. Lancet Neurol. 2010;9:1070–7.

    Article  CAS  PubMed  Google Scholar 

  178. Stockner H, Iranzo A, Seppi K, Serradell M, Gschliesser V, Sojer M, et al. Midbrain hyperechogenicity in idiopathic REM sleep behavior disorder. Mov Disord. 2009;24:1906–9.

    Article  PubMed  Google Scholar 

  179. Iwanami M, Miyamoto T, Miyamoto M, Hirata K, Takada E. Relevance of substantia nigra hyperechogenicity and reduced odor identification in idiopathic REM sleep behavior disorder. Sleep Med. 2010;11:361–5.

    Article  PubMed  Google Scholar 

  180. Shin HY, Joo EY, Kim ST, Dhong H-J, Cho JW. Comparison study of olfactory function and substantia nigra hyperechogenicity in idiopathic REM sleep behavior disorder, Parkinson’s disease and normal control. Neurol Sci. 2013;34:935–40.

    Article  PubMed  Google Scholar 

  181. Unger MM, Belke M, Menzler K, Heverhagen JT, Keil B, Stiasny-Kolster K, et al. Diffusion tensor imaging in idiopathic REM sleep behavior disorder reveals microstructural changes in the brainstem, substantia nigra, olfactory Region, and other brain Regions. Sleep. 2010;33:767–73.

    Article  PubMed  PubMed Central  Google Scholar 

  182. Ahmadi S-A, Bötzel K, Levin J, Maiostre J, Klein T, Wein W, et al. Analyzing the co-localization of substantia nigra hyper-echogenicities and iron accumulation in Parkinson’s disease: a multi-modal atlas study with transcranial ultrasound and MRI. Neuroimage Clin. 2020;26: 102185.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Li D-H, He Y-C, Liu J, Chen S-D. Diagnostic accuracy of transcranial sonography of the substantia Nigra in Parkinson’s disease: a systematic review and meta-analysis. Sci Rep-uk. 2016;6:20863.

    Article  CAS  Google Scholar 

  184. Heller J, Brcina N, Dogan I, Holtbernd F, Romanzetti S, Schulz JB, et al. Brain imaging findings in idiopathic REM sleep behavior disorder (RBD) – A systematic review on potential biomarkers for neurodegeneration. Sleep Med Rev. 2017;34:23–33.

    Article  PubMed  Google Scholar 

  185. Wilson H, Dervenoulas G, Pagano G, Tyacke RJ, Polychronis S, Myers J, et al. Imidazoline 2 binding sites reflecting astroglia pathology in Parkinson’s disease: an in vivo11C-BU99008 PET study. Brain. 2019;142:3116–28.

    Article  PubMed  Google Scholar 

  186. Ferini-Strambi L, Oertel W, Dauvilliers Y, Postuma RB, Marelli S, Iranzo A, et al. Autonomic symptoms in idiopathic REM behavior disorder: a multicentre case–control study. J Neurol. 2014;261:1112–8.

    Article  PubMed  Google Scholar 

  187. Miyamoto T, Miyamoto M, Inoue Y, Usui Y, Suzuki K, Hirata K. Reduced cardiac 123I-MIBG scintigraphy in idiopathic REM sleep behavior disorder. Neurology. 2006;67:2236–8.

    Article  CAS  PubMed  Google Scholar 

  188. McDade EM, Boot BP, Christianson TJH, Pankratz VS, Boeve BF, Ferman TJ, et al. Subtle gait changes in patients with REM sleep behavior disorder. Movement Disord. 2013;28:1847–53.

    Article  PubMed  Google Scholar 

  189. Giannini G, Provini F, Cortelli P, Calandra-Buonaura G. REM sleep behaviour disorder in multiple system atrophy: from prodromal to progression of disease. Front Neurol. 2021;12:677213.

    Article  PubMed  PubMed Central  Google Scholar 

  190. Wilke C, Santos MCT, Schulte C, Deuschle C, Scheller D, Verbelen M, et al. Intraindividual neurofilament dynamics in serum mark the conversion to sporadic Parkinson’s disease. Mov Disord. 2020;35:1233–8.

    Article  CAS  PubMed  Google Scholar 

  191. Sommer U, Hummel T, Cormann K, Mueller A, Frasnelli J, Kropp J, et al. Detection of presymptomatic Parkinson’s disease: combining smell tests, transcranial sonography, and SPECT. Mov Disord. 2004;19:1196–202.

    Article  PubMed  Google Scholar 

  192. Siderowf A, Newberg A, Chou KL, Lloyd M, Colcher A, Hurtig HI, et al. [99mTc]TRODAT-1 SPECT imaging correlates with odor identification in early Parkinson disease. Neurology. 2005;64:1716–20.

    Article  CAS  PubMed  Google Scholar 

  193. Jennings D, Siderowf A, Stern M, Seibyl J, Eberly S, Oakes D, et al. Imaging prodromal Parkinson disease. Neurology. 2014;83:1739–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Fernández-Santiago R, Iranzo A, Gaig C, Serradell M, Fernández M, Tolosa E, et al. MicroRNA association with synucleinopathy conversion in rapid eye movement behavior disorder. Ann Neurol. 2015;77:895–901.

    Article  PubMed  Google Scholar 

  195. Baumgartner U, Berger F, Gheinani AH, Burgener SS, Monastyrskaya K, Vassella E. miR-19b enhances proliferation and apoptosis resistance via the EGFR signaling pathway by targeting PP2A and BIM in non-small cell lung cancer. Mol Cancer. 2018;17:44.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Horai T, Boku S, Okazaki S, Otsuka I, Ratta-apha W, Mouri K, et al. miR-19b is elevated in peripheral blood of schizophrenic patients and attenuates proliferation of hippocampal neural progenitor cells. J Psychiatr Res. 2020;131:102–7.

    Article  PubMed  Google Scholar 

  197. Smith AM, Depp C, Ryan BJ, Johnston GI, Alegre-Abarrategui J, Evetts S, et al. Mitochondrial dysfunction and increased glycolysis in prodromal and early Parkinson’s blood cells. Movement Disord. 2018;33:1580–90.

    Article  CAS  PubMed  Google Scholar 

  198. Heintz-Buschart A, Pandey U, Wicke T, Sixel-Döring F, Janzen A, Sittig-Wiegand E, et al. The nasal and gut microbiome in Parkinson’s disease and idiopathic rapid eye movement sleep behavior disorder. Mov Disord. 2017;6:e28032.

    Google Scholar 

  199. Fairfoul G, McGuire LI, Pal S, Ironside JW, Neumann J, Christie S, et al. Alpha-synuclein RT-QuIC in the CSF of patients with alpha-synucleinopathies. Ann Clin Transl Neur. 2016;3:812–8.

    Article  CAS  Google Scholar 

  200. Bargar C, Wang W, Gunzler SA, LeFevre A, Wang Z, Lerner AJ, et al. Streamlined alpha-synuclein RT-QuIC assay for various biospecimens in Parkinson’s disease and dementia with Lewy bodies. Acta Neuropathol Commun. 2021;9:62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Liu X, Yang J, Yuan Y, He Q, Gao Y, Jiang C, et al. Optimization of the detection method for phosphorylated α-synuclein in Parkinson disease by skin biopsy. Front Neurol. 2020;11:569446.

    Article  PubMed  PubMed Central  Google Scholar 

  202. Rossi M, Candelise N, Baiardi S, Capellari S, Giannini G, Orrù CD, et al. Ultrasensitive RT-QuIC assay with high sensitivity and specificity for Lewy body-associated synucleinopathies. Acta Neuropathol. 2020;140:49–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Stefani A, Iranzo A, Holzknecht E, Perra D, Bongianni M, Gaig C, et al. Alpha-synuclein seeds in olfactory mucosa of patients with isolated REM sleep behaviour disorder. Brain. 2021;144(4):1118–26.

    Article  PubMed  Google Scholar 

  204. Donadio V, Wang Z, Incensi A, Rizzo G, Fileccia E, Vacchiano V, et al. In vivo diagnosis of synucleinopathies: a comparative study of skin biopsy and RT-QuIC. Neurology. 2021. https://doi.org/10.1212/WNL.0000000000011935.

    Article  PubMed  PubMed Central  Google Scholar 

  205. Doty RL. Olfactory dysfunction in Parkinson disease. Nat Rev Neurol. 2012;8:329–39.

    Article  CAS  PubMed  Google Scholar 

  206. Ross GW, Petrovitch H, Abbott RD, Tanner CM, Popper J, Masaki K, et al. Association of olfactory dysfunction with risk for future Parkinson’s disease. Ann Neurol. 2008;63:167–73.

    Article  PubMed  Google Scholar 

  207. Postuma RB, Iranzo A, Hu M, Högl B, Boeve BF, Manni R, et al. Risk and predictors of dementia and parkinsonism in idiopathic REM sleep behaviour disorder: a multicentre study. Brain. 2019;142:awz030.

    Article  Google Scholar 

  208. Berg D, Marek K, Ross GW, Poewe W. Defining at-risk populations for Parkinson’s disease: lessons from ongoing studies. Mov Disord. 2012;27:656–65.

    Article  PubMed  Google Scholar 

  209. Domellöf ME, Lundin K-F, Edström M, Forsgren L. Olfactory dysfunction and dementia in newly diagnosed patients with Parkinson’s disease. Parkinsonism Relat D. 2017;38:41–7.

    Article  Google Scholar 

  210. Cecchini MP, Federico A, Zanini A, Mantovani E, Masala C, Tinazzi M, et al. Olfaction and taste in Parkinson’s disease: the association with mild cognitive impairment and the single cognitive domain dysfunction. J Neural Transm. 2019;126:585–95.

    Article  PubMed  Google Scholar 

  211. Marrero-González P, Iranzo A, Bedoya D, Serradell M, Niñerola-Baizán A, Perissinotti A, et al. Prodromal Parkinson disease in patients with idiopathic hyposmia. J Neurol. 2020;267:3673–82.

    Article  PubMed  Google Scholar 

  212. Silveira-Moriyama L, Holton JL, Kingsbury A, Ayling H, Petrie A, Sterlacci W, et al. Regional differences in the severity of Lewy body pathology across the olfactory cortex. Neurosci Lett. 2009;453:77–80.

    Article  CAS  PubMed  Google Scholar 

  213. Poulopoulos M, Levy OA, Alcalay RN. The neuropathology of genetic Parkinson’s disease. Mov Disord. 2012;27:831–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Burke RE, Dauer WT, Vonsattel JPG. A critical evaluation of the Braak staging scheme for Parkinson’s disease. Ann Neurol. 2008;64:485–91.

    Article  PubMed  PubMed Central  Google Scholar 

  215. Halliday G, Hely M, Reid W, Morris J. The progression of pathology in longitudinally followed patients with Parkinson’s disease. Acta Neuropathol. 2008;115:409–15.

    Article  PubMed  Google Scholar 

  216. Kalaitzakis ME, Graeber MB, Gentleman SM, Pearce RKB. The dorsal motor nucleus of the vagus is not an obligatory trigger site of Parkinson’s disease: a critical analysis of α-synuclein staging. Neuropathol Appl Neurobiol. 2008;34:284–95.

    Article  CAS  PubMed  Google Scholar 

  217. Ulusoy A, Rusconi R, Pérez-Revuelta BI, Musgrove RE, Helwig M, Winzen-Reichert B, et al. Caudo-rostral brain spreading of α-synuclein through vagal connections. Embo Mol Med. 2013;5:1119–27.

    Article  PubMed  Google Scholar 

  218. Ulusoy A, Phillips RJ, Helwig M, Klinkenberg M, Powley TL, Monte DAD. Brain-to-stomach transfer of α-synuclein via vagal preganglionic projections. Acta Neuropathol. 2017;133:381–93.

    Article  CAS  PubMed  Google Scholar 

  219. Arotcarena M-L, Dovero S, Prigent A, Bourdenx M, Camus S, Porras G, et al. Bidirectional gut-to-brain and brain-to-gut propagation of synucleinopathy in non-human primates. Brain. 2020;143:1462–75.

    Article  PubMed  Google Scholar 

  220. Borghammer P. How does parkinson’s disease begin? Perspectives on neuroanatomical pathways, prions, and histology. Mov Disord. 2018;33:48–57.

    Article  PubMed  Google Scholar 

  221. Knudsen K, Fedorova TD, Hansen AK, Sommerauer M, Otto M, Svendsen KB, et al. In-vivo staging of pathology in REM sleep behaviour disorder: a multimodality imaging case-control study. Lancet Neurol. 2018;17:618–28.

    Article  PubMed  Google Scholar 

  222. Horsager J, Andersen KB, Knudsen K, Skjærbæk C, Fedorova TD, Okkels N, et al. Brain-first versus body-first Parkinson’s disease: a multimodal imaging case-control study. Brain. 2020;143:3077–88.

    Article  PubMed  Google Scholar 

  223. Schulz-Schaeffer WJ. Is cell death primary or secondary in the pathophysiology of idiopathic Parkinson’s disease? Biomol. 2015;5:1467–79.

    CAS  Google Scholar 

  224. Lang AE, Siderowf AD, Macklin EA, Poewe W, Brooks DJ, Fernandez HH, et al. Trial of Cinpanemab in Early Parkinson’s Disease. New Engl J Med. 2022;387:408–20.

    Article  CAS  PubMed  Google Scholar 

  225. Pagano G, Taylor KI, Anzures-Cabrera J, Marchesi M, Simuni T, Marek K, et al. Trial of prasinezumab in early-stage Parkinson’s disease. New Engl J Med. 2022;387:421–32.

    Article  CAS  PubMed  Google Scholar 

  226. Whone A. Monoclonal antibody therapy in Parkinson’s disease — The End? New Engl J Med. 2022;387:466–7.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

N.a

Funding

Open Access funding enabled and organized by Projekt DEAL. N.a.

Author information

Authors and Affiliations

Authors

Contributions

T.K. and S.L.R. wrote the first draft of the manuscript; P.S., F.S., M.B., J.L., C.T., G.U.H. & J.H. revised and edited the manuscript; T.K. prepared the final version of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Thomas Koeglsperger.

Ethics declarations

Ethics approval and consent to participate

N.a.

Consent for publication

All authors consented to publish the manuscript

Competing interests

We declare that we have no competing interests. J.L. reports speaker fees from Bayer Vital, Biogen, EISAI, TEVA and Roche, consulting fees from Axon Neuroscience and Biogen, author fees from Thieme medical publishers and W. Kohlhammer GmbH medical publishers and is inventor in a patent “Oral Phenylbutyrate for Treatment of Human 4-Repeat Tauopathies” (EP 23 156 122.6) filed by LMU Munich. In addition, he reports compensation for serving as chief medical officer for MODAG GmbH, is beneficiary of the phantom share program of MODAG GmbH and is inventor in a patent “Pharmaceutical Composition and Methods of Use” (EP 22 159 408.8) filed by MODAG GmbH, all activities outside the submitted work.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Koeglsperger, T., Rumpf, SL., Schließer, P. et al. Neuropathology of incidental Lewy body & prodromal Parkinson’s disease. Mol Neurodegeneration 18, 32 (2023). https://doi.org/10.1186/s13024-023-00622-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13024-023-00622-7

Keywords