Skip to main content
Molecular Neurodegeneration logoLink to Molecular Neurodegeneration
. 2025 Nov 3;20:117. doi: 10.1186/s13024-025-00900-6

Neuropathology of Lewy body dementia: Lewy-related pathology, α-synuclein oligomers, and comorbid pathologies

Hiroaki Sekiya 1,, Tomoyasu Matsubara 2, Michael A DeTure 1, Dennis W Dickson 1,
PMCID: PMC12581614  PMID: 41185066

Abstract

Lewy body dementia is the second most common form of neurodegenerative dementia, following Alzheimer’s disease. This umbrella term encompasses dementia with Lewy bodies (DLB) and Parkinson’s disease dementia (PDD). The distinction between these two conditions lies in the timing of the onset of cognitive impairment relative to motor symptoms. In DLB, cognitive impairment precedes or coincides with motor symptoms within the first year, whereas in PDD, cognitive decline occurs more than a year after the onset of motor symptoms. Clinically, in addition to cognitive decline, patients with Lewy body dementia have parkinsonism, visual hallucinations, and fluctuations of cognitive status. The pathological hallmark of this condition is the presence of Lewy bodies and Lewy neurites, collectively referred to as Lewy-related pathology. This is identical to Parkinson’s disease, where dementia is not observed. The principal component of Lewy-related pathology is α-synuclein, which classifies this disorder as an α-synucleinopathy. While Lewy-related pathology represents a later stage of α-synuclein aggregation, earlier stages involve α-synuclein oligomers. Emerging evidence suggests α-synuclein oligomers may be more toxic than Lewy-related pathology. In addition to α-synuclein pathology, previous studies frequently observed comorbid pathological conditions, including Alzheimer’s disease neuropathologic change, TAR DNA-binding protein 43 (TDP-43) pathology, and cerebral small vessel disease among others. In this review, we provide a comprehensive overview of the underlying pathologies for Lewy body dementia and their molecular mechanisms and clinical implications. We also discuss concepts including the prion-like propagation hypothesis of α-synuclein, α-synuclein strain hypothesis, and recent advances in machine learning algorithms for analyzing propagation patterns. The purpose of this manuscript is to elucidate these complex pathological conditions, advance our understanding of the disease, and improve diagnostic strategies.

Supplementary Information

The online version contains supplementary material available at 10.1186/s13024-025-00900-6.

Keywords: Lewy body dementia, Dementia with Lewy bodies, Parkinson’s disease dementia, Alpha-synuclein, Oligomers, Comorbid pathology, Prion-like propagation, Machine learning algorithms, Neuropathology, Lewy bodies

Introduction

Dementia with Lewy bodies (DLB) is the second most common form of neurodegenerative dementia after Alzheimer’s disease (AD). Clinical manifestations of this condition include recurrent visual hallucinations, fluctuations in cognitive status, REM sleep behavior disorder (RBD), and parkinsonism, in addition to cognitive decline [1]. Parkinson’s disease (PD) is a movement disorder that presents with extrapyramidal symptoms including bradykinesia, rigidity, and tremor. In addition to these motor symptoms, increasing attention has been focused on nonmotor symptoms, including cognitive dysfunction [2, 3]. Indeed, dementia prevalence increases over the course of PD, with more than 80% of individuals surviving 20 years developing dementia [4]. This condition is referred to as Parkinson’s disease dementia (PDD). “Lewy body dementia” is the collective term for DLB and PDD. The one-year rule distinguishes DLB and PDD; DLB is diagnosed when the patient presents with dementia before or within one year after the onset of parkinsonism, while PDD is diagnosed when the patient develops dementia more than one year after the onset of parkinsonism. If patients never develop dementia, the diagnosis is PD.

The pathological hallmark of Lewy body dementia is eosinophilic intraneuronal inclusions called Lewy bodies. Lewy bodies are observed in the cytoplasm of neurons and are mainly composed of pathological aggregates of α-synuclein [5, 6]. Morphologically, Lewy bodies include brainstem-type with a characteristic halo and cortical-type without a distinct halo. Pathological aggregates of α-synuclein are also detected in dystrophic neurites, called Lewy neurites. Collectively, Lewy bodies and Lewy neurites are referred to as Lewy-related pathology. While Lewy-related pathology represents a late-stage aggregate composed of α-synuclein fibrils, α-synuclein forms oligomers consisting of several molecules in the early phase of its aggregation, which may be in equilibrium with α-synuclein fibrils [7, 8]. Recently, there has been growing interest in the potential neurotoxicity of these early-stage α-synuclein aggregates [911]. Furthermore, the frequent co-occurrence of other pathological conditions in Lewy body dementia has been described, including the accumulation of non-α-synuclein protein aggregates and vascular pathology [1214].

This review provides a comprehensive overview of the underlying neuropathology in Lewy body dementia, ranging from the conventional Lewy-related pathology to α-synuclein oligomers and comorbid pathologies, and suggests directions for future research.

Terminology

In this field, inconsistent use of terms leads to confusion in the terminology [15]. In particular, the abbreviation “LBD” is used for both Lewy body disease and Lewy body dementia, further compounding this confusion. Therefore, we begin this review by reaffirming the definitions of each term. While the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5) has replaced “dementia” with “major neurocognitive disorder” [16], terms such as “Lewy body dementia,” “dementia with Lewy bodies,” and “Parkinson’s disease dementia” continue to be widely used in clinical practice and research. For consistency with current literature, we use these established terms throughout this review, while acknowledging the evolving nature of diagnostic terminology. Similarly, definitions of “cognitive impairment” and “dementia” may vary across studies cited in this review. We have generally preserved original terminology used in each research paper while acknowledging heterogeneity in diagnostic criteria and assessment methods. Therefore, the threshold distinguishing cognitive impairment from dementia may differ between studies due to methodological differences.

Figure 1 illustrates the terminology associated with α-synucleinopathy. Pathologically, neurodegenerative disorders characterized by pathological aggregation of α-synuclein are collectively referred to as α-synucleinopathies [17]. Lewy body disease is a major category of α-synucleinopathies characterized by the presence of Lewy bodies, which are α-synuclein-positive spherical inclusions in the neuronal cytoplasm [5]. Lewy body disease encompasses DLB, PD, PDD, and pure autonomic failure. These disorders share the common pathological feature of Lewy bodies but differ in their clinical presentation and progression. Another significant α-synucleinopathy is multiple system atrophy (MSA), characterized by widespread and abundant glial cytoplasmic inclusions [18, 19]. Glial cytoplasmic inclusions are α-synuclein-positive triangular or oval inclusions found in the oligodendroglial cytoplasm [20]. Although neurons also contain α-synuclein aggregates [2123], these glial cytoplasmic inclusions remain the pathological hallmark.

Fig. 1.

Fig. 1

Terminology of α-synucleinopathy. DLB, dementia with Lewy bodies; MSA, multiple system atrophy; PAF, pure autonomic failure; PD, Parkinson’s disease; PDD, Parkinson’s disease dementia; RBD, REM sleep behavior disorder

Patients with DLB present with cognitive impairment along with parkinsonism, visual hallucinations, RBD, and cognitive fluctuations. In contrast, patients with PD primarily exhibit extrapyramidal motor symptoms and a variety of non-motor symptoms. Although patients with PD do not initially present with cognitive impairment, a certain proportion develop dementia over time [24], and their condition is subsequently categorized as PDD. DLB and PDD are collectively referred to as Lewy body dementia.

It is important to note that while PD, PDD, DLB, and Lewy body dementia are clinical terms used for diagnosis based on clinical presentation, Lewy body disease is a pathological term. Lewy body disease refers to the presence of Lewy bodies in the brain, which can be definitively confirmed only through post-mortem examination. Lewy body disease is further classified into diffuse Lewy body disease, transitional Lewy body disease, and brainstem-predominant Lewy body disease based on the distribution of Lewy bodies. Generally, brainstem-predominant Lewy body disease corresponds to clinical PD, while transitional and diffuse Lewy body disease are more commonly associated with clinical DLB, although clinicopathological correlations can be complex.

In this review, while the abbreviations PD, PDD, and DLB will be used, the full spelling will be used for Lewy body disease and Lewy body dementia instead of using abbreviations to avoid confusion.

Clinical and pathological spectrum of PDD and DLB

PDD and DLB are diagnosed based on the one-year rule in clinical practice as described above. The relationship between these two conditions has been debated; some researchers consider them as distinct entities and others view them as different manifestations of the same disease.

Table 1 summarizes the similarities and differences in clinical features between PDD and DLB. Many of the clinical differences between PDD and DLB naturally reflect their diagnostic definitions – DLB presents with early cognitive symptoms while motor symptoms are the initial manifestation in PDD. However, the pattern of disease progression appears to show more intrinsic differences beyond these definition-based distinctions. For instance, DLB patients tend to show more severe postural instability and gait difficulty despite having less severe parkinsonism overall. Additionally, they often experience more frequent falls early in the disease course compared to PDD patients. While both conditions eventually manifest similar symptoms, these differences in disease progression suggest potential differences in disease mechanisms.

Table 1.

Similarities and differences in clinical features between PDD and DLB

Clinical symptoms Similarities Differences
Bradykinesia Core feature of parkinsonism (slowness of movement and difficulty initiating movements)

Prevalence: cardinal symptom in nearly all PDD; present in up to 87% of DLB [25]

Severity: Higher at baseline in PDD [26]

Progression: Associated with more rapid decline in DLB [26]

Tremor Can present as a resting tremor (rhythmic, occurs when the affected limb is at rest)

Type: Mixed pattern (resting and postural/action) can be seen in DLB [27] Prevalence: Common in PDD; variable in DLB (25–74%) [25, 27, 28]

Progression: Lower risk of developing dementia in tremor-dominant PD [29, 30]

Rigidity Common parkinsonian feature (increased muscle tone and resistance to passive movement) Prevalence: Common in PDD; 48% in DLB [26]
Postural instability, recurrent falls Implications for disease progression and patient mobility [31, 32]; Associated with higher risk of developing dementia in PD [33]

Prevalence: 48–73% in PDD [34, 35]; 35–40% in DLB [25, 34]

Diagnostic value: Falls in 5 years preceding diagnosis more strongly associated with DLB [36]

RBD Onset: Often precedes other symptoms [3742] Impact: associated with more severe disease course [38, 41, 43, 44] Prevalence: higher in DLB (50–90%) than PDD (likely similar to or slightly higher than PD; 25–58%) [3941, 45]
Visual hallucinations

Complex, well-formed, unpleasant hallucinations [46]

Associated with worse outcomes (more rapid cognitive decline, increased mortality, higher caregiver burden, increased risk of nursing home placement) [47, 48]

Prevalence: Higher in DLB vs PDD (72% vs 50% [49], 63% vs 20% [50], 51% vs 29% [51], 32% vs 11% [48]); 62% in DLB by meta-analysis in DLB [52]
Delusions Associated with more rapid cognitive decline and increased caregiver burden [51]

Prevalence: Higher in DLB vs PDD (57% vs 29% [49], 52% vs 29% [51])

Severity: Often more pronounced in DLB [51]

Cognitive fluctuations Severity: Comparable fluctuating attention [53] Prevalence: Higher in DLB (25%) vs PDD (9%) [50]
Cognitive profiles Comparable visuospatial abilities at baseline and progression [54]

More severe memory impairment in DLB and more severe executive impairment in PDD at baseline; Faster decline in executive function in PDD and faster decline in language ability in DLB [54]

Worse inhibitory control and semantic fluency in DLB [55]

Orthostatic hypotension

Prevalence: 49% in PDD, 52% in DLB [56]

Prognosis: Associated with shorter survival [57]

Urinary incontinence Associated with poor prognosis [57] Prevalence: 50–97% in DLB [5860]; in PDD, likely similar to that in PD (30% by meta-analysis [61])

Table 2 summarizes the pathological similarities and differences between PDD and DLB. Although findings are not always consistent across studies, DLB generally shows greater pathological burden compared to PDD in terms of α-synuclein, tau, amyloid β, and cerebral amyloid angiopathy. The discrepancies between studies may be attributed to variations in antibodies used and methods for evaluating pathological burden.

Table 2.

Similarities and differences in pathological findings between PDD and DLB

References Subjects Brain regions Methods Findings
Similarities
Harding et al. (2001) [62] 25 PD, 16 PDD, 24 DLB Cortex (frontal, temporal, cingulate, and parahippocampal) LB counts on α-synuclein IHC (LB509) Comparable LB counts in cortex
Tsuboi et al. (2005) [63] 13 PD, 6 PDD, 28 DLB Cortex (frontal, temporal, parietal, cingulate and parahippocampal), hippocampus, and amygdala LB counts on NACP-IHC at x200 magnification, SP and NFT counts on thioflavin S Comparable LB counts in cortex and amygdala, SP counts in cortex and hippocampus, and NFT counts in hippocampus
Tsuboi et al. (2007) [64] 7 PD, 7 PDD, 14 DLB Putamen % stained area of α-synuclein IHC Comparable % stained area in putamen
Kalaitzakis et al. (2009) [65] 20 PD, 12 PDD, 7 DLB Claustrum Semi-quantitative assessment of Aβ (4G8) IHC Comparable Aβ in claustrum
Kalaitzakis et al. (2011) [66] 52 PD, 41 PDD, 14 DLB, 7 MSA, 12 PSP Putamen and caudate nucleus Semi-quantitative assessment of Aβ (4G8) IHC Comparable Aβ in putamen and caudate nucleus
Sierra et al. (2016) [67] 10 PD, 10 PDD, 10 DLB, 10 AD, 10 Ctrl Midbrain and Cerebellum Semi-quantitative assessment of α-synuclein (KM51), p-tau (AT8), and Aβ (6F/3D) IHC Similar α-synuclein, Aβ, and p-tau burden in midbrain and comparable Aβ burden in cerebellum
Differences
Jellinger et al. (2006) [68] 17 PDD, 17 DLB Striatum Semi-quantitative assessment of LRP, p-tau (AT8), and Aβ (4G8) IHC A greater burden of LRP, Aβ plaques and tau pathology in striatum in DLB
Kalaitzakis et al. (2009) [65] 20 PD, 12 PDD, 7 DLB Claustrum Semi-quantitative assessment of α-synuclein (BD) IHC A greater α-synuclein burden in claustrum in DLB
Halliday et al. (2011) [69] 7 PD, 12 PDD, 10 DLB, 5 ctrl Striatum Semi-quantitative assessment of Aβ (IE8) IHC A greater burden of striatal Aβ plaques in DLB
Ruffmann et al. (2015) [70] 49 PD, 55 PDD, 17 DLB Cortex, striatum Semi-quantitative assessment of α-synuclein (42 or 211) and Aβ (4G8) IHC A greater α-synuclein burden in temporal and parietal cortices and greater Aβ burden in striatum and frontal cortex in DLB
Hepp et al. (2016) [71] 27 PD, 56 PDD, 50 DLB Cortex, amygdala, striatum % stained area of Aβ (6F/3D) IHC A greater Aβ burden in frontal, temporal, occipital, entorhinal cortices and caudate nucleus in DLB
Jellinger (2021) [72] 60 PD, 110 PDD, 60 DLB Cortex Semi-quantitative assessment of CAA severity with Aβ (4G8) IHC More severe CAA in DLB
Jellinger (2023) [73] 100 PD, 110 PDD, 80 DLB Cortex Semi-quantitative assessment of CAA severity with Aβ (4G8) IHC More severe CAA in DLB

Aβ, amyloid β; AD, Alzheimer’s disease; Ctrl, control: DLB, dementia with Lewy bodies; IHC, immunohistochemistry; LB, Lewy body; LRP, Lewy-related pathology; MSA, multiple system atrophy; NFT, neurofibrillary tangle; PD, Parkinson’s disease; PDD, Parkinson’s disease dementia; PSP, progressive supranuclear palsy; p-tau, phosphorylated tau; SP, senile plaque

Although the one-year rule is somewhat arbitrary, it is useful and widely used as a classification in clinical practice and in research. This distinction helps in the differential diagnosis and management of PDD and DLB, which, despite their overlap, may have different therapeutic and prognostic implications. For example, DLB often presents with higher amyloid and tau pathology compared to PDD [74]. In research, maintaining this distinction aids in the stratification of study populations, ensuring more homogeneous groups for clinical trials and epidemiological studies. This can improve the validity and reliability of research findings and facilitate the development of targeted therapies [75].

Epidemiology

DLB is now recognized as one of the most common causes of dementia, affecting up to 5% of the general population and accounting for up to 31% of patients with dementia. The incidence of DLB is estimated to be 0.5–1.6 per 1,000 person-years in the general population and 32–38 per 1,000 person-years in patients with dementia [7678]. Regarding PDD, a systematic review revealed that PDD accounts for 25–31% of patients with PD and represents 3.2–5.3% of all patients with dementia [24].

The wide ranges observed may be attributed to differences in diagnostic criteria, study methodologies, and population characteristics across studies. It should be noted that not all epidemiological studies differentiate PDD and DLB. Furthermore, the accurate diagnosis of Lewy body dementia in clinical practice remains challenging [79, 80].

A study of clinicopathologic correlations in 273 consecutive autopsies of nursing home residents reported that Lewy bodies were found in 17% [81]. In a combination of two cohorts, the Rush Religious Order Study and the Memory and Aging Project, 18% of 872 autopsies had Lewy bodies [82]. A prospective cohort study from Hisayama town in Japan showed that among 1,266 autopsies (with an approximate autopsy rate of 75% in the town), the prevalence of DLB was 4.4% in the general population and 14.4% in patients with dementia, respectively [83].

The prevalence of dementia in PD increases over the disease course, with approximately 80% of PD patients developing dementia (i.e., PDD) after 20 years from motor symptom onset [4]. Given the increasing prevalence of PD [84], the prevalence of PDD is expected to rise in the future.

Genetics of Lewy body dementia

Lewy body dementia is characterized by several genetic features, which have been identified through various genetic association studies. The most consistently replicated genetic associations involve mutations in the APOE, GBA1, and SNCA genes [85].

The APOE gene encodes apolipoprotein E and is polymorphic with three major alleles: ε2, ε3, and ε4 [86, 87]. The APOE ε4 allele is a significant genetic risk factor for Lewy body dementia and influences the disease’s progression and Lewy-related pathology [8891]. Although the APOE ε4 is the strongest risk factor for AD [9295], a study of 652 autopsy-confirmed Lewy body disease demonstrated that patients with APOE ε4 had higher Lewy body burden in the cortices, independent of AD pathology [96]. Furthermore, in a study involving 81 PDD, 91 DLB without AD pathology, 224 Lewy body disease with AD pathology, 244 AD, and 269 control cases, the frequency of APOE ε4 was significantly higher in PDD and DLB without AD pathology compared to controls [97]. While many studies have shown that APOE ε4 is associated with Lewy body dementia, it should be noted that a recent study did not confirm the involvement of APOE ε4 in DLB without AD pathology [98]. Clinically, a study of 3,923 patients with PD showed that 6.7% converted to PDD during the observation period, and that APOE ε4 was associated with the development of dementia [99]. Several studies have also reported that APOE ε4 carriers with PD or Lewy body dementia experience more rapid cognitive decline [100103]. Experimental studies using mouse models also suggest the association between APOE ε4 and α-synuclein pathology. One study showed that APOE ε4 increased α-synuclein pathology and impaired behavioral performance independently of amyloid pathology [104]. In a study using SNCA A53T transgenic mice, mice on APOE ε4 showed more abundant phosphorylated α-synuclein pathology in the brainstem compared to APOE knockout, ε2, ε3 mice, and had more α-synuclein accumulation in the substantia nigra pars compacta than ε2 or APOE knockout mice [105].

The GBA1 gene encodes the lysosomal enzyme, glucocerebrosidase. While homozygous mutations in the GBA1 gene cause Gaucher disease, heterozygous mutations in the GBA1 gene are a significant risk factor for PD and Lewy body dementia [106]. Many studies have demonstrated the association between GBA1 gene variants and Lewy body dementia [90, 107111]. Reduced glucocerebrosidase levels contribute to increased α-synuclein accumulation in Lewy body dementia, partly through elevating glucosylsphingosine levels, ultimately impacting disease manifestation [112]. Clinical characteristics of Lewy body dementia with GBA1 mutations include younger onset, more frequent and severe parkinsonism, and higher frequency of visual hallucinations and fluctuations [113]. Liu et al. screened 1,921 patients with PD for GBA1 mutations and found that carriers of neuropathic Gaucher disease mutations and complex GBA1 alleles exhibited a more rapid decline in Mini-Mental State Examination (MMSE) scores, whereas this association was not observed in carriers of non-neuropathic mutations or risk variants [114]. Mata et al. analyzed 1,369 patients with PD for GBA1 mutations and reported that both GBA1 mutations and the E326K polymorphism were associated with poorer performance in working memory/executive function and visuospatial abilities [115]. Pathologically, one study revealed an inverse relationship between the frequency of GBA1 mutations/polymorphisms and the severity of comorbid AD in Lewy body dementia [116], suggesting that GBA1 genetic variants may influence the development of Lewy body dementia independent of comorbid AD. A recent study using 943 autopsy-confirmed Lewy body disease found that those with GBA1 mutations exhibited lower Braak neurofibrillary tangle stage and Thal amyloid phase and more severe Lewy-related pathology [117].

The SNCA gene, which encodes α-synuclein, is another critical genetic factor in Lewy body dementia [85, 88]. A point mutation in the SNCA gene (A53T) was first reported in a large pedigree of familial PD [118, 119]. Since then, other point mutations including V15A, A30P/G, E46K, H50Q, and G51D, A53E/V, T72M, E83Q, as well as duplications, and triplications have been described [120136]. The pathologic findings of SNCA point mutations based on postmortem human brain samples are summarized in Table 3. Many point mutations show more severe pathology compared to sporadic PD, and some mutations have been reported to exhibit glial cytoplasmic inclusions. Of note, these point mutations in SNCA have been reported to promote oligomer formation of α-synuclein (α-synuclein oligomers will be discussed in more detail later) [142145]. A study with 198 PDD and 922 DLB patients identified novel SNCA gene variations associated with PDD (rs10018362, rs7689942) and DLB (rs974711, rs1348224), with rs1348224 showing association in both conditions, suggesting both shared and distinct genetic risk factors within SNCA for these disorders [146]. A meta-analysis study using data from 59 families with SNCA multiplications revealed that SNCA multiplications were associated with the development of Lewy body dementia and that higher SNCA copy numbers correlated with earlier onset of cognitive impairment [147]. As for cognitive function, a recent study compared 16 patients with PD carrying the SNCA A53T mutation to age- and disease duration-matched idiopathic PD patients. While Montreal Cognitive Assessment score was comparable at baseline, the A53T group demonstrated greater cognitive decline over the three-year follow-up period. Moreover, executive and visuospatial functions were poorer in the A53T group at baseline compared to idiopathic PD and further deteriorated during disease progression [148].

Table 3.

Pathological features of patients with SNCA point mutations

Mutations References AAD Sex Duration Description
A30P Seidel et al. (2010) [126] 69 M 15

Neuronal loss in SNpc, LC, dmX

Widespread LRP

Glial inclusions

E46K Zarranz et al. (2004) [124] 69 M 8

Neuronal loss in dmX and SNpc

Widespread LRP

Microspongiosis in SNpc

A53E Pasanen et al. (2014) [131] 60 F 24

Neuronal loss in SNpc, hippocampus, cortices (temporal, insular, cingulate)

A few LBs in dmX

Neuronal and glial cytoplasmic inclusions in putamen, caudatus, amygdala, hippocampus, cortices (temporal, insular, cingulate)

A53T Golbe et al. (1990) [118] and Duda et al. (2002) [137] 49 M 11

Neuronal loss in SNpc, hippocampus

LBs in SNpc, LC, dmX, raphe nucleus, nbM, limbic cortices

LNs in LC, dmX, amygdala, CA2, CA3, parahippocampal gyrus, striatum, motor cortex

A53T Golbe et al. (1990) [118] 64 M 4

Neuronal loss in LC

LBs in LC

*Only cerebral cortex, basal ganglia, pons, cerebellum, and cervical cord available

A53T Spira et al. (2001) [138] 47 M 5

Neuronal loss in SNpc, LC, CA2, CA3

LBs in SNpc and LC

LNs in SNpc, LC, dmX, CA2, CA3, cortices

Vacuolation in temporal cortex

A53T Spira et al. (2001) [138] 55 M 9

Neuronal loss in SNpc, LC, CA2, CA3, putamen, globus pallidus, temporal cortex

LBs in SNpc, LC, oculomotor nucleus

LNs in SNpc, LC, dmX, oculomotor nucleus

No glial inclusions

A53T Yamaguchiet al. (2005) [139] 50 F 9

Neuronal loss in dmX, SNpc, nbM, limbic, cortex

LBs in LC, amygdala, hippocampus, temporal cortex; more widespread and abundant LNs

Microvacuolation in nbM, nucleus accumbens, cortices (entorhinal, cingulate, temporal)

A53T Markopoulou et al. (2008) [140] 71 F 12

Neuronal loss in SNpc, LC, nbM, amygdala, CA1, subiculum, temporal cortex

LRP in dmX, LC, SNpc, nbM, hypothalamus, limbic cortices

A53T Markopoulou et al. (2008) [140] 39 M 7

Neuronal loss in SNpc, nbM, CA2

LRP in dmX, LC, SNpc, nbM, amygdala, limbic cortices

Glial cytoplasmic inclusions in limbic and temporal cortices

A53T Nishioka et al. (2020) [141] 52 M 10

Neuronal loss in SNpc, LC, dmX

Widespread Lewy-related pathology

LB

Mild glial cytoplasmic inclusions in amygdala, hippocampus, cortices (frontal, temporal, parietal, occipital)

H50Q Proukakis et al. (2013) [129] and Kiely et al. (2015) [130] 83 F 12

Neuronal loss in SNpc

LRP in SNpc, putamen, caudate nucleus, cortex (Braak PD stage 6)

No glial inclusions

G51D Kiely et al. (2013) [127] and Kiely et al. (2015) [130] 48 M 19

Neuronal loss in SNpc, LC, dmX, CA2, CA3, cortices (temporal and insular)

Various morphology of neuronal α-synuclein cytoplasmic inclusions in wide regions including cortices

Glial cytoplasmic inclusions

G51D Lesage et al. (2013) [127] 67 F 7

Neuronal loss in SNpc, LC, putamen, caudate nucleus, globus pallidus, thalamus

LBs in SNpc, LC, dmX, hippocampus, cingulate cortex

LNs in spinal cord, SNpc, LC, putamen, caudate nucleus, globus pallidus, hippocampus, cortices (frontal, cingulate, motor)

Annular or crescent neuronal cytoplasmic inclusions in spinal cord, LC, dmX, hippocampus, cortices (cingulate, motor)

G51D Kiely et al. (2015) [130] 75 ND 6

Neuronal loss in SNpc, LC, dmX, CA2, CA3, cingulate cortex

Various morphology of neuronal α-synuclein cytoplasmic inclusions in wide regions including cortices

Glial cytoplasmic inclusions

G51D Kiely et al. (2015) [130] 52 ND 6

Neuronal loss in SNpc, LC, dmX

Various morphology of neuronal α-synuclein cytoplasmic inclusions in wide regions including cortices

Glial cytoplasmic inclusions

E83Q Kapasi et al. (2020) [133] 63 F 4

Neuronal loss in SNpc, amygdala, cortices (entorhinal, cingulate, temporal)

LBs in SNpc, amygdala, basal ganglia, thalamus, CA1, cortices (entorhinal, cingulate, temporal, frontal, parietal)

Spongiform changes in cortices (entorhinal, cingulate, temporal)

No autopsy report was found for patients with SNCA V15A, A30G, A53V, or T72M

AAD, age at death; dmX, dorsal motor nucleus of the vagus; F, female; LBs, Lewy bodies; LC, locus coeruleus; LNs, Lewy neurites; LRP, Lewy-related pathology; M, male; nbM, nucleus basalis of Meynert; ND, no data; SNpc, substantia nigra parc compacta

Furthermore, SNCB has also been linked to Lewy body dementia. β-synuclein, encoded by SNCB, inhibits α-synuclein aggregation [149] and α-synuclein oligomer formation [150], and cortical β-synuclein levels are reduced in diffuse Lewy body disease without comorbid AD pathology [151]. Specific insertion and deletion mutations in the SNCB gene are frequently observed in DLB, potentially contributing to the decrease of β-synuclein [152]. Conversely, β-synuclein accumulation is observed in dopaminergic neurons [153], and mutations such as V70M and P123H in SNCB [154] are associated with DLB and exacerbate neurodegeneration [155].

Additionally, correlations between MAPT haplotypes and Lewy body dementia have been documented. The MAPT H1/H1 haplotype is associated with more severe synuclein burden [156]. While the MAPT H2 haplotype has a protective effect, a relatively rare H1G haplotype is linked to an increased risk of DLB [157]. The rare H1j haplotype significantly correlates with neurodegeneration in the putamen [158].

Lewy-related pathology

The pathological hallmark of PD and DLB is Lewy bodies and Lewy neurites, which are collectively referred to as Lewy-related pathology [159163]. Lewy bodies are round inclusions in cytoplasm of neurons, while Lewy neurites are aggregations in the dystrophic neurites. Lewy bodies are ultrastructurally assemblies of filamentous and amorphous granular material [164], the major component of which is α-synuclein consisting of 140 amino acid residues [5], with phosphorylation at serine 129 [165]. While fibrillar α-synuclein forms the structural backbone of Lewy bodies, studies with immunohistochemistry [166170] and proteomic analyses of isolated Lewy bodies [171, 172] have consistently demonstrated that ubiquitin, p62/SQSTM1, and phosphorylated neurofilament subunits also constitute Lewy bodies, reflecting activation of protein quality control pathways and cytoskeletal pathways. Furthermore, these proteomic analyses have catalogued over 100 additional proteins associated with Lewy bodies, highlighting that Lewy bodies are complex protein assemblies whose complete functional hierarchy remains to be elucidated.

Macroscopic pathology

In Lewy body dementia, the brain typically does not exhibit significant atrophy (Fig. 2A), and the brain weight remains within normal limits or only slightly lower than normal. If atrophy or a marked decrease in brain weight is observed, other diagnoses or the influence of co-pathologies such as comorbid AD should be considered (Fig. 2B, C). In the brainstem, Lewy body dementia shares the characteristic feature of depigmentation of both the substantia nigra and the locus coeruleus with PD (Fig. 2D, E). The substantia nigra is particularly susceptible in the ventrolateral and caudal segments [173]. In addition, atrophy or discoloration of the basal ganglia is usually not observed in Lewy body dementia.

Fig. 2.

Fig. 2

Macroscopic images. Lateral view of the brain with dementia with Lewy bodies shows no significant atrophy (A). Coronal brain sections at the level of the lateral geniculate body showing the hippocampus: Lewy body dementia without Alzheimer’s disease neuropathological changes (B) and Lewy body dementia with Alzheimer’s disease neuropathological changes (C). Substantia nigra and locus coeruleus in dementia with Lewy bodies shows loss of neuromelanin pigment (D, F) in contrast with a normal subject (E, G). Scale bars: 2 cm (A) and 0.5 cm (B-G)

Microscopic pathology

The histological hallmark of Lewy body dementia is Lewy bodies [174]. The classic Lewy body, mainly observed in the brainstem, is an intraneuronal cytoplasmic inclusion consisting of a dense and concentric hyaline eosinophilic core surrounded by a halo (Fig. 3A). Similar inclusions are also observed within neuronal processes, particularly in the dorsal motor nucleus of the vagal nerve and the peripheral sympathetic nerve, and are termed intraneuritic Lewy bodies [174, 175] (Fig. 3B). Less dense, mildly eosinophilic inclusions without a halo are referred to as pale bodies, often observed in the pigmented neurons of the substantia nigra, and are considered an immature form of Lewy bodies [176] (Fig. 3C). In the cortex, smaller and round intraneuronal eosinophilic cytoplasmic inclusions without a distinct halo are observed and termed cortical Lewy bodies [177] (Fig. 3D). Lewy bodies observed in the brainstem are typically relatively large, spherical, and strongly eosinophilic, characterized by a dense central core and a surrounding halo. In contrast, cortical Lewy bodies are relatively smaller, less intensely eosinophilic, and usually lack a halo. Despite these morphological differences, both types of Lewy bodies can be visualized using immunohistochemistry for α-synuclein (Fig. 3E, F) and ubiquitin [166]. Immunostaining for α-synuclein also allows the detection of Lewy neurites, which are not visible with hematoxylin and eosin staining [178] (Fig. 3G). Lewy neurites are generally observed more abundant than Lewy bodies in pathological sections. This is primarily due to their structural differences: Lewy neurites represent α-synuclein accumulation within long processes, whereas Lewy bodies are inclusions within the neuronal soma. Given that histological sections are typically thin (5–10 µm), elongated processes are more likely to be captured in any given plane of sectioning, while spherical soma may be missed depending on the cutting angle. Additionaly, although Lewy neurites appear more abundant than Lewy bodies, the burden of both pathologies tends to increase in nearly parallel as the disease progresses [179, 180]. Therefore, Lewy neurites and Lewy bodies are considered fundamentally the same pathology differing only in morphology. Immunohistochemistry has uncovered that Lewy bodies are widely distributed in both the central and peripheral nervous systems [181184]. Microvacuolation of the neuropil is another characteristic of Lewy body dementia [185, 186], which is anatomically confined mainly to the limbic cortices, especially the amygdala and entorhinal cortex (Fig. 3H). Ultrastructural studies using electron microscopy have shown that these vacuoles are located within dendrites and/or axons, suggesting that they reflect degeneration of these neuronal processes [185, 187, 188]. This change occurs independently of any prion disease-related mechanisms, as immunohistochemistry with antibodies against prion protein failed to label plaques or vascular amyloid deposits [185]. The pattern and distribution of spongiform change in Lewy body disease also differ from those in prion diseases such as Creutzfeldt-Jakob disease, where spongiform change is widespread, confluent, and a core diagnostic criterion [189].

Fig. 3.

Fig. 3

Microscopic images of Lewy body dementia. (A) brainstem Lewy bodies in the substantia nigra; (B) intraneuritic Lewy body in the dorsal motor nucleus of the vagal nerve; (C) pale body in the substantia nigra; (D) cortical Lewy bodies in the cingulate cortex; (E) brainstem Lewy body in the substantia nigra; (F) cortical Lewy bodies in the cingulate gyrus; (G) numerous Lewy neurites in the CA2 subfield of the hippocampus; (H) spongiform change in the pre-α layer of the entorhinal cortex. (A-D, H): hematoxylin and eosin staining, (E-G): immunohistochemistry for phosphorylated α-synuclein. Scale bars: 20 µm (A-F), 50 µm (G, H)

Distribution of Lewy-related pathology

Lewy body disease can be classified based on the distribution pattern of Lewy bodies [1, 190] (Table 4). Kosaka et al. classified Lewy body disease into several categories: diffuse, transitional, and brainstem-predominant [190]. Diffuse Lewy body disease is the pathological diagnostic term for cases in which Lewy bodies are widely distributed in the neocortex, while brainstem-predominant Lewy body disease is used when Lewy bodies are mainly confined to the brainstem with minimal distribution in the cerebrum. Transitional Lewy body disease is used for cases in which the distribution of Lewy bodies is intermediate between the two, and the Lewy bodies are found in the limbic system, which is also referred to as limbic Lewy body disease. Braak et al. classified PD into 6 stages based on the extent of Lewy body distribution, ranging from minimally affected brains to those with widespread involvement [181] (Table 4). A group of patients was reported with amygdala-predominant Lewy bodies without notable Lewy bodies in the brainstem [192, 193]. These subtypes are often observed as a concomitant alteration in AD [193]. Moreover, AD with Lewy bodies often exhibits the coexistence of tau and α-synuclein in the same neuron [194196] (comorbid AD will be discussed in detail below). While various staging schemes have been proposed [179, 183, 191, 197199], the current pathological classification of Lewy body disease into five types is now widely used: diffuse (neocortical), transitional (limbic), brainstem-predominant, amygdala-predominant, and olfactory bulb-only [1]. In diffuse Lewy body disease, brainstem Lewy-related pathology is present in nearly all patients, to some extent. Even in individuals without clinically overt parkinsonism, Lewy-related pathology and neuronal loss in the substantia nigra pars compacta are often observed. This discrepancy can be explained by the “threshold effect”, which suggests that approximately 50–90% loss of nigral dopaminergic (tyrosine hydroxylase-positive) neurons is required for the manifestation of motor symptoms [200]. The dissociation between pathological changes and clinical symptoms in DLB may be attributed to neuronal loss below this threshold, precedence of cognitive symptoms, or different progression patterns of nigral pathology.

Table 4.

Staging of Lewy body disease

Kosaka stage [190] Braak PD stage [181] Unified staging system [191] Distribution of Lewy bodies
- 1 Olfactory bulb only (group I) Olfactory bulb
Brainstem-predominant type (group C) Brainstem-predominant type (group IIa) Brainstem and limbic type (group III) Medullary tegmentum and the dorsal motor nucleus of the vagal nerve
2 Locus coeruleus, caudal raphe and reticular nucleus of medullary and pontine tegmentum
3 Substantia nigra and basal forebrain
Transitional (limbic) type (group B) 4 Limbic predominant type (group IIb) Medial temporal (limbic) cortices, including amygdala and CA2 region of the hippocampus
Diffuse cortical type (group A) 5 Neocortical type (group IV) Multimodal association cortices, especially frontal and temporal lobes
6 Unimodal association cortices and primary cortices

PD, Parkinson’s disease; CA, cornu ammonis

In early or incidental Lewy body disease, the distribution of Lewy bodies is often confined to the brainstem and peripheral autonomic nerves or to the olfactory bulb and amygdala [201, 202]; in advanced Lewy body disease, the distribution of Lewy bodies is more widespread throughout the brain, giving rise to the “propagation hypothesis” discussed below.

In addition to the distribution of Lewy-related pathology in the central nervous system, Lewy-related pathology is also observed in the peripheral nervous system, particularly in areas closely associated with autonomic function, such as the nerve plexus in the walls of the esophagus and intestines [182, 183, 203205], submandibular gland [206, 207], nerve fibers in the skin [208210], paraspinal sympathetic ganglia and sympathetic nerves surrounding the heart [184, 203, 211, 212], adrenal glands [213, 214], kidney and genitourinary tract [182, 215], and retina [216].

Molecular mechanisms of neurodegeneration

Multiple molecular mechanisms are proposed to underlie neurodegeneration associated with Lewy-related pathology. First, loss of normal α-synuclein function results in decreased synaptic plasticity [217, 218], impaired neurotransmitter release [219, 220], abnormal SNARE complex formation [221, 222], vesicular transport defects [223, 224], and disrupted dopamine synthesis and transport [225227]. Second, toxic gain-of-function mechanisms from abnormal α-synuclein aggregate accumulation cause axonal transport impairment [228, 229] and inhibition of protein degradation systems, including the lysosomal-autophagy [230, 231] and ubiquitin-proteasome pathways [232, 233]. Additionally, direct damage to intracellular organelles manifests as mitochondrial dysfunction [234236], lysosomal membrane rupture [237, 238], and cytoskeletal disruption [239, 240]. Furthermore, prion-like mechanisms of intercellular transmission and aggregate spreading via extracellular vesicles contribute to pathological progression (pathology propagation will be discussed in detail later) [241, 242]. These complex mechanisms might interact synergistically, ultimately leading to neuronal death.

However, the relationship between Lewy-related pathology and neurodegeneration may be more complex than previously thought. Emerging evidence suggests that α-synuclein oligomers, which are though to precede Lewy-related pathology formation [243], exhibit greater toxicity than mature aggregates (α-synuclein oligomers will be discussed later) [9, 244]. This has led to the “protective aggregate” hypothesis, proposing that Lewy-related pathology formation may represent a cellular defense mechanism that sequesters highly toxic α-synuclein oligomers [245247]. Supporting this concept, a Drosophila PD model demonstrated that inhibiting α-synuclein phosphorylation at serine 129 increased Lewy body-like inclusions while paradoxically suppressing dopamine neuron loss, whereas mimicking phosphorylation decreased inclusions but enhanced neurotoxicity [248]. Dynamic modeling and in silico studies have demonstrated that early anti-oligomer intervention provides neuroprotective effects while late-stage inhibition of aggregate formation proves counterproductive, and that rapid fibril formation from α-synuclein oligomers minimizes neuronal toxicity [249, 250]. These findings suggest a potential neuroprotective function of aggregate formation, although further investigation is needed to fully understand this complex relationship.

Clinical implication of Lewy-related pathology

A large number of clinicopathological studies have been conducted on the impact of Lewy-related pathology on clinical manifestations. In one study using a prospectively followed cohort with 49 DLB patients, among α-synuclein, tau, and amyloid β, α-synuclein contributed most to reduced disease duration [251]. Another study also demonstrated that diffuse Lewy body disease had shorter disease duration compared to transitional Lewy body disease [252].

Regarding the overall distribution of Lewy-related pathology and clinical symptoms, studies have examined the correlation between pathological distribution based on the Unified Staging System for Lewy Body disorders and symptoms [253]. Among 280 patients, 9% were classified as stage 1 (olfactory bulb only), 15% as stage IIa (brainstem predominant), 14% as stage IIb (limbic predominant), 32% as stage III (brainstem and limbic), and 31% as stage IV (neocortical). Correlations between these stages and parkinsonism, cognitive impairment, RBD, and olfactory dysfunction have been reported.

Cognitive impairment

The association between dementia and Lewy-related pathology has been investigated by examining differences in the degree of Lewy-related pathology between PD and PDD or DLB. An autopsy study including 20 PD and 22 PDD patients revealed that the severity of neocortical Lewy-related pathology was higher in PDD [254]. Another study with 25 PD, 16 PDD, and 24 DLB patients reported that the amount of Lewy-related pathology in the parahippocampal gyrus was most useful in differentiating PD from PDD or DLB [62]. Comparison between 9 PD and 12 PDD patients demonstrated that the number of Lewy bodies was much higher in the amygdala, limbic cortex, and neocortex in PDD [255]. In a study with 60 PD and 69 PDD patients, cortical Lewy body scores were significantly higher in PDD [256]. Furthermore, the severity of cortical Lewy-related pathology was higher in PDD in a study with 48 PD and 92 PDD patients [257], and a higher cortical Lewy body score contributed most to dementia in a study involving 46 PD and 109 PDD patients [258]. Regarding the distribution of Lewy-related pathology, PDD has a higher proportion of transitional or diffuse patterns compared to PD without dementia [255, 258].

In contrast, there are several reports indicating that cortical Lewy-related pathology is not necessarily associated with dementia. For example, 19% of 16 PDD patients or 24% of 34 PDD patients exhibited Lewy-related pathology confined to the brainstem [259, 260]. Another study examined the distribution pattern of Lewy-related pathology in 17 PD patients and 21 PDD patients and reported that the neocortical form was observed in 47% of PD patients and 67% of PDD patients, with no significant difference [261]. A large retrospective clinicopathological study of 106 autopsy patients revealed that only 20% of patients with cortical Lewy-related pathology were clinically diagnosed with PDD or DLB [262]. In addition to cortical pathology, a recent study compared the severity of Lewy-related pathology in the midbrain and reported that the amount of Lewy-related pathology in the substantia nigra and midbrain tectum/tegmentum was not different among PD, PDD, and DLB [67].

In summary, while several studies indicate a higher severity of cortical Lewy-related pathology in PDD and DLB compared to PD, other reports suggest that cortical Lewy-related pathology alone may not be a definitive marker for dementia. These findings highlight the complexity of the relationship between Lewy-related pathology and dementia. Lewy bodies in the substantia nigra were observed in a constant proportion of neurons in PD and DLB regardless of symptoms or disease duration, suggesting that Lewy bodies are removed at the death of neurons harboring them [263]. This phenomenon may complicate the correlation between Lewy-related pathology and clinical manifestations. Furthermore, comorbid conditions including amyloid and tau (discussed in detail below) may contribute to cognitive impairment.

Regarding the severity of cognitive impairment and Lewy-related pathology, a clinicopathological study with 44 PDD patients demonstrated that severity of cognitive impairment (Reisberg’s global deterioration scale) correlated with the number of cortical Lewy bodies [264]. Furthermore, the same authors showed that the number of Lewy bodies in the cingulate gyrus, frontal gyrus, temporal gyrus, amygdala, straight gyrus, and angular gyrus correlated with the severity of cognitive impairment [265]. Another study with 88 consecutive autopsies of PD or PDD (9 without cognitive impairment and 79 with cognitive impairment) revealed that the Mini-Mental State Examination score correlated with the distribution of Lewy-related pathology (i.e. Braak PD stage) [266]. Additionally, it was also reported that the severity of Lewy-related pathology in PD correlated with the final score on the Mini-Mental State Examination [267] and the rate of its decline [268]. Using another dementia scale, one study demonstrated a correlation between the density of Lewy-related pathology in the cortex and the nucleus basalis of Meynert and the clinical dementia rating [81].

These studies suggest that the degree of Lewy-related pathology may affect the severity of cognitive impairment in Lewy body disease; however, the brain regions that contribute to this relationship vary between studies. These differences may be attributed to the retrospective nature of many of the studies with varying degrees of accuracy of clinical information and the limited number of patients involved. Future clinicopathologic studies with a large number of prospectively followed-up patients are warranted.

Visual hallucinations

There are also studies that have explored clinicopathological correlations for symptoms other than cognitive impairment. Core clinical features of DLB include recurrent visual hallucinations, cognitive fluctuations, RBD, and parkinsonism [1]. Recurrent complex visual hallucinations are frequently observed in patients with Lewy body dementia and the prevalence of visual hallucinations in DLB is about 60% [46, 52]. Patients with PDD and DLB experience well-formed hallucinations including people or animals [46]. The underlying pathology causing visual hallucinations is thought to be Lewy-related pathology rather than tau [269272]. A study with 129 autopsies of PD (69 with dementia) examined the severity of cortical Lewy pathologies and revealed that patients with visual hallucinations had a greater cortical Lewy pathology severity [256]. Another pathological study examined the correlation between visual hallucinations and Lewy body burden in individual brain regions and revealed that patients with visual hallucinations exhibited a greater number of Lewy bodies in the parahippocampal cortex and amygdala [273]. Interestingly, when patients were divided by the time of onset of visual hallucinations, those with early-onset visual hallucinations had more Lewy bodies in the parahippocampal gyrus and temporal lobe, whereas those with late-onset of visual hallucinations showed no difference from those without visual hallucinations. This suggests a time lag between the onset of visual hallucinations and the formation of Lewy-related pathology. Another study investigated Lewy-related pathology in 19 autopsied brains of patients with DLB, 14 of whom had visual hallucinations [274]. Although the secondary visual pathway and amygdala showed more severe Lewy-related pathology than the primary visual pathway, no significant differences in pathological findings were observed between patients with and without visual hallucinations. These studies highlight the complex relationship between visual hallucinations and the distribution of Lewy-related pathology in DLB and warrant future research.

Cognitive fluctuations

Fluctuations in DLB include waxing and waning of cognition, functional abilities, and arousal, and such fluctuations were observed in 63% of DLB patients [275]. A clinicopathological study demonstrated that patients with a distribution of Lewy bodies in the neocortex were more prone to fluctuations in their cognition, although the fluctuations were assessed using variation across annual average cognitive decline instead of hourly- or daily-based evaluations [82]. Additionally, functional imaging studies have suggested that the thalamus [276278], basal ganglia [277, 279, 280], parietal lobe [279, 281, 282], occipital lobe [276, 281], or frontoparietal connections [283, 284] are potentially responsible for fluctuations in cognitive status [285, 286]. These fluctuations, however, are dynamic phenomena of very short duration, and pathological validation of the responsible regions remains elusive.

REM sleep behavior disorder

During REM sleep, the normal muscle state is characterized by atonia, which is an almost complete paralysis of the skeletal muscles. RBD is a parasomnia with dream-enactment behaviors caused by a loss of REM sleep muscle atonia [287, 288]. The involvement of the brainstem nuclei in the pathogenesis of RBD has been suggested [289291]. Patients with α-synucleinopathy often present with RBD long before the onset of motor symptoms or cognitive decline [1, 292294]. Regarding the distribution of α-synuclein pathology, a study demonstrated that PD patients with RBD exhibited more severe α-synuclein burden in many brain regions, including the brainstem and cortices, compared to those without RBD [295]. Additionally, PD patients with RBD had a higher positive rate of phosphorylated α-synuclein in colon biopsies compared to those without RBD, suggesting that PD patients with RBD have a more widespread distribution of α-synuclein pathology [296]. Attempts have been made to identify the responsible foci of RBD by pathology, however, no convincing results have been obtained at present. A neuropathological study showed that cholinergic neuron counts in the pedunculopontine and laterodorsal tegmental nuclei were comparable between DLB and MSA patients with and without RBD [297]. Another study examined neuronal loss and α-synuclein burden in the pedunculopontine/laterodorsal tegmental nucleus and locus coeruleus between Lewy body disease patients with and without RBD, which found no difference [298]. Recently, in addition to cholinergic neurons [299, 300], the involvement of GABAergic and glutamatergic neurons has been suggested as a possible pathogenesis of RBD [301305]. While Lewy-related pathology has been observed in GABAergic and glutamatergic neurons [306308], the correlation between these findings and RBD requires further investigation. Additionally, studies have suggested the involvement of orexin neurons in RBD [309, 310]; however, orexin neurons are generally considered to lack Lewy-related pathology despite its presence in the hypothalamus [311]. Therefore, further research is needed to determine whether RBD and orexin neurons are indeed related in α-synucleinopathy.

Other clinical manifestations

Clinicopathological studies have also been conducted on other manifestations of Lewy body dementia including daytime sleepiness and psychiatric symptoms. In a study examining sleep patterns in 61 DLB and 26 AD patients, DLB patients frequently demonstrated shorter initial sleep latency on multiple sleep latency tests and higher Epworth Sleepiness Scale scores [312]. Furthermore, among 20 DLB patients who underwent autopsy, 12 were classified as diffuse Lewy body disease and 8 as transitional Lewy body disease, suggesting that excessive daytime sleepiness can occur even in the absence of cortical Lewy bodies. Conversely, an analysis of 211 autopsy cases from the Honolulu-Asia Aging Study revealed that while 17.5% (20/114) of patients without Lewy-related pathology exhibited excessive daytime sleepiness, this symptom was present in 43.9% (25/57) of patients with neocortical Lewy-related pathology (Braak PD stage ≥ 5), demonstrating a correlation between the distribution of Lewy-related pathology and daytime hypersomnolence [313].

Regarding psychiatric symptoms, several studies have demonstrated associations between the distribution of Lewy-related pathology and clinical manifestations. In a study of 112 DLB patients, persistent delusions were found to increase with more widespread Lewy body distribution, occurring in 11% of brainstem-predominant, 32% of transitional, and 37% of diffuse Lewy body disease cases [270]. Another study revealed a greater number of Lewy bodies in the dorsal raphe nucleus in Lewy body disease with delusion compared to those without delusion [314]. A recent study using National Alzheimer’s Coordinating Center Registry data, which included 658 cases of Lewy body disease, demonstrated an increased frequency of delusions, anxiety, depression, and apathy as Lewy-related pathology spread from the brainstem to limbic structure and neocortex [315].

Additionally, a small number of clinicopathologic studies has been performed on atypical symptoms, including aphasia and behavioral changes. A large brain bank study identified 11 diffuse Lewy body disease patients presenting with corticobasal syndrome among 523 autopsy-confirmed diffuse Lewy body disease patients, and showed that these patients had more Lewy bodies in the motor cortex than those with clinically probable DLB [316]. Patients with diffuse Lewy body disease who clinically presented with logopenic progressive aphasia had a greater number of Lewy bodies in the middle frontal cortex, inferior parietal cortex, and anterior cingulate gyrus compared to those diffuse Lewy body disease patients clinically presenting with DLB [317]. These studies suggest that not only the type of accumulated protein but also where that protein accumulates may influence clinical presentation.

Propagation hypothesis of pathological α-synuclein

Braak et al. conducted immunostaining for α-synuclein in autopsy cases of PD and clinically asymptomatic “incidental” Lewy body disease to reveal a gradient of pathological “spread” from the brainstem to the cerebrum. Based on this observation, they hypothesized that Lewy-related pathology propagates through the nervous system [181, 318, 319]. This study established a paradigm for subsequent research by translating the spatial distribution of lesions into a temporal axis. This propagation hypothesis was supported by several reports demonstrating that α-synuclein accumulation was observed within fetal nigral tissue transplanted into PD patients [320322]. Additionally, experimental replication of α-synuclein propagation in the brain was achieved by inoculating synthetic α-synuclein preformed fibrils or seeds extracted from the brains of PD patients into the basal ganglia of mice [323, 324] and non-human primates [325, 326]. In recent years, studies in animals have reported the propagation of Lewy-related pathology to the cerebrum and brainstem after injection of preformed fibrils or seeds into the gastrointestinal tract [327, 328] or olfactory bulb [329332].

In most cases of Lewy body disease without comorbid pathologies, Lewy-related pathology follows a stereotypical ascending pattern from the brainstem to the cerebrum, as confirmed by a number of studies replicating Braak’s hypothesis [333]. However, in Lewy body disease with comorbid AD, Lewy-related pathology in the amygdala is found from an early stage and even in the absence of Lewy-related pathology in the brainstem [192, 193, 334]. This underscores the heterogeneity of the propagation process.

Origin of pathological α-synuclein

Given that pathological α-synuclein propagates and spreads throughout the brain, then where does the pathological α-synuclein originate? Braak et al. proposed that the gastrointestinal tract as the origin of pathological α-synuclein [319]. This proposal was based on their observation that the dorsal motor nucleus of the vagus nerve and/or the olfactory bulb are the sites where Lewy-related pathology is detected in the least affected brain, as well as on the findings that Lewy bodies are observed in the enteric nervous system [335, 336]. Since the vagus nerve, which innervates the gut, retrogrades to enter the dorsal motor nucleus of the vagus nerve in the medulla, it was hypothesized that the pathological α-synuclein originating in the gut could enter the central nervous system via the vagus nerve. Epidemiological data indicating a lower risk of developing PD in individuals who have undergone vagotomy [337, 338] along with the detection of Lewy bodies in intestinal specimens collected before the onset of motor symptoms [204, 339342], are considered circumstantial evidence supporting the gastrointestinal origin of Lewy-related pathology. In contrast, there have been no cases where Lewy-related pathology was detected exclusively in the gastrointestinal tract [343345], whereas several cases have been reported in which Lewy-related pathology was observed only in the olfactory bulb [183, 191, 346], cardiac sympathetic nerves [183, 347], or adrenal glands [213]. These cases suggest that the origin of Lewy-related pathology may be other than the gastrointestinal tract. Other hypotheses have been proposed, including the olfactory bulb as the origin [191], both the gastrointestinal tract and the olfactory bulb as origins [348355], or a multicentric origin [356361]. Each of these hypotheses lacks conclusive evidence or is refuted by contrary evidence.

Prion-like propagation mechanisms

Experimental evidence from both in vitro and in vivo models demonstrates prion-like propagation of α-synuclein [242, 362365]. Luk et al. demonstrated that introducing exogenous recombinant α-synuclein fibrils into cells overexpressing α-synuclein induces the recruitment of endogenous α-synuclein and the formation of Lewy body-like inclusions [366]. Furthermore, extracellular α-synuclein oligomers can cross cell membranes and trigger intracellular aggregation of endogenous α-synuclein in both primary neurons and neuronal cell lines [367]. Aulić et al. showed that a single exposure of SH-SY5Y cells to recombinant α-synuclein amyloid fibrils could induce aggregation of endogenous wild-type α-synuclein, even without overexpression and that the seeded aggregates persisted and propagated over several cell passages [368]. In vivo models also provide evidence for α-synuclein prion-like propagation. Desplats et al. demonstrated direct α-synuclein transmission by showing inclusion body formation in mouse cortical neural stem cells stereotactically injected into the hippocampus of transgenic mice that overexpress human α-synuclein [369]. In another study, intracerebral injection of brain homogenates from symptomatic α-synuclein transgenic mice or recombinant α-synuclein fibrils into young, asymptomatic α-synuclein transgenic mice rapidly induced widespread Lewy-related pathology-like inclusions, leading to motor symptoms and reduced survival [370]. Furthermore, stereotactic injection of insoluble α-synuclein from brains of patients with DLB and recombinant α-synuclein fibrils into wild-type mouse brains resulted in abnormal phosphorylation of endogenous mouse α-synuclein and its time- and region-dependent propagation from the injection site [324].

Disease-specific propagation patterns

Notably, multiple studies using patient brain homogenates have demonstrated distinct α-synuclein propagation properties among synucleinopathies, including PD, DLB, and MSA. In vitro studies revealed fundamental differences in seeding properties: MSA-derived α-synuclein exhibits diffuse inclusion-forming capacity in both soluble and insoluble fractions with more efficient seeding activity, whereas PD-derived α-synuclein shows localized inclusion-forming capacity only in the insoluble fraction [371]. Cell-based assays using α-synuclein mutant HEK cells demonstrated distinct aggregation patterns, with MSA inducing large aggregates in most cell lines except for E46K, DLB forming small aggregates in all cell lines, and PD producing very small aggregates [372]. In vivo propagation experiments have consistently demonstrated the greater pathogenic potential of MSA-derived α-synuclein across different mouse models. Studies using α-synuclein A53T transgenic mice showed that MSA brain homogenate induced severe motor impairment and widespread α-synuclein pathology, while DLB brain homogenate produced no neurological symptoms and only localized pathological changes [373, 374]. Similarly, experiments in wild-type mice demonstrated that MSA brain homogenate induced more severe and widespread α-synuclein pathology with stronger microglial activation compared to PD brain homogenate [375]. Early studies confirmed that MSA brain tissue induced neurodegeneration in both cell and transgenic mouse models, while PD brain tissue did not [376]. Another study using wild-type rats further validated these findings, showing that MSA-derived α-synuclein assemblies are more aggressive and potent in inducing neuropathological and behavioral phenotypes compared to PD and DLB assemblies [377]. These findings collectively support the “α-synuclein strain hypothesis” that proposes disease-specific prion-like propagation mechanisms among synucleinopathies.

Computational approaches to propagation analysis

Recent advances in computational pathology have provided new insights into α-synuclein propagation patterns. Pathological data obtained from postmortem brains represent single time points at death for each patient, making it challenging to track progression of pathology through brain regions over time within the same patient. To address this limitation, the Subtype and Stage Inference (SuStaIn) algorithm enables probabilistic reconstruction of temporal disease progression patterns [378, 379] from various types of cross-sectional data such as neuroimaging [380389] and neuropathology [390393]. Several studies have explored pathological progression patterns by applying the SuStaIn algorithm to Lewy-related pathology data. Mastenbroek et al. utilized semi-quantitative scores across ten brain regions in 814 autopsy cases, identifying three distinct progression patterns [391]. In 82% of cases, Lewy-related pathology initially accumulated in the olfactory bulb, with subsequent involvement of either the limbic system (61% of total cases) or brainstem (21% of total cases). The remaining 18% exhibited initial pathology in the brainstem. Another study by Andersen et al. applied the algorithm to two datasets (173 and 129 cases) [393]. With notable inclusion of many cases with incidental Lewy body disease and assessment of peripheral tissue pathology, their analysis identified brain-first (51%) and body-first (49%) subtypes, with the latter subdivided into parasympathetic and sympathetic variants. These approaches may enable the extraction of temporal data through pseudo-time analysis from static, single-timepoint pathological data, potentially revealing disease progression patterns.

Although the hypothesis of pathological α-synuclein propagation has been widely studied and supported by experimental evidence, the origin of pathological α-synuclein remains to be elucidated. Advancing our understanding of the origins and pathways of α-synuclein propagation will be crucial in developing effective interventions for Lewy body dementia.

α-synuclein oligomer and its clinical implication

Toxic mechanisms of α-synuclein oligomers

In the protein aggregation process, the intermediates from monomers to fibrils are termed oligomers, which are transient, heterogeneous higher-order protein aggregates [394] (Fig. 4A). In α-synucleinopathies, while Lewy-related pathology is the late-stage aggregates of α-synuclein, early-stage α-synuclein aggregates, known as α-synuclein oligomers, are attracting more attention in the pathogenesis. Mounting evidence has demonstrated that such α-synuclein oligomers are more toxic than Lewy-related pathology [395, 396]. Various mechanisms have been proposed for the toxicity of oligomeric species of synuclein based on cellular and animal studies. Among these toxicity pathways, membrane disruption is considered the primary [395, 397406]. A recent study demonstrated that α-synuclein oligomers had a higher affinity for the membrane compared to monomeric α-synuclein and that they replace monomers on the membrane [406]. Other targets include mitochondrial dysfunction [407412], synaptic impairment [401, 407, 413419], endoplasmic reticulum stress [420423], and oxidative stress [396, 424]. Additionally, several studies have suggested that α-synuclein oligomers elicit an inflammatory response, resulting in neuronal damage [424428].

Fig. 4.

Fig. 4

α-synuclein oligomers. (A) The aggregation process of α-synuclein. Multiple molecules form an early-stage aggregate of α-synuclein, known as an α-synuclein oligomer. (B) an anti-α-synuclein antibody conjugated with one oligonucleotide (PLUS) and an anti-α-synuclein antibody conjugated with another oligonucleotide (MINUS) are applied to the tissue. These antibodies bind in close proximity only in the presence of α-synuclein oligomers. This proximity allows subsequent reactions (ligation and amplification) to occur, enabling the visualization of α-synuclein oligomers

Detection methods for α-synuclein oligomers

Methods for detecting α-synuclein oligomers include enzyme-linked immunosorbent assays [429431], Förster resonance energy transfer-based techniques [432], and nanopore-based single-molecule detection [433]. These approaches can detect α-synuclein oligomers with high sensitivity in samples such as biological fluids, including cerebrospinal fluid, blood, and brain homogenates, as well as cell lysates. However, none of these methods preserve or provide information on the topographical distribution of α-synuclein oligomers within brain regions. Immunohistochemistry using oligomer-specific antibodies has been attempted to visualize α-synuclein oligomers in pathological sections; however, a systematic validation study has shown that widely used conformation-specific or oligomer-specific monoclonal antibodies bind not only to α-synuclein oligomers but also to fibrils and monomers [434]. This cross-reactivity limits the ability of immunohistochemistry to distinguish between oligomers and other aggregated forms of α-synuclein in tissue. To address this issue, a recent technique has enabled visualization of α-synuclein oligomers on histopathological slides. Proximity Ligation Assay (PLA) was originally developed as a method to detect endogenous protein interactions [435]. Fig. 4B illustrates how this method detects α-synuclein oligomers. By combining antigen-antibody reactions with in situ DNA amplification techniques, target proteins can be detected with high sensitivity and specificity. Two types of oligonucleotides are conjugated to antibodies against two target proteins, and only when the target proteins are in close proximity (within 40 nm), the two oligonucleotides are ligated, creating a template for amplification. This template is then amplified by rolling circle amplification. Complementary oligonucleotides labeled with horseradish peroxidase hybridize to the amplified product, and the signal is visualized by adding a substrate. This method was applied to detect α-synuclein oligomers by binding two different oligonucleotides to the same α-synuclein antibody [22, 428, 436440]. This α-synuclein-PLA staining detected α-synuclein oligomers in morphologically normal neurons. Such α-synuclein oligomers were found in brain regions that were mildly affected [436]. Moreover, two independent research groups have successively reported that α-synuclein oligomers are distributed even in patients with LRRK2-associated PD who did not have Lewy-related pathology [441, 442]. The application of α-synuclein-PLA staining may reveal early α-synuclein pathology that has not been previously visualized. Some studies suggest that endogenous α-synuclein exists as helically folded tetramers [443, 444], while other research indicates it exists as disordered monomers [445]. Previous experiments using α-synuclein PLA staining did not observe many signals in control subjects [22, 436, 439, 441, 442, 446, 447]. Theoretically, α-synuclein PLA staining visualizes α-synuclein oligomers by recognizing two or more α-synuclein molecules in close proximity, typically within 40 nm. Physiological tetramers are tightly folded structures resistant to aggregation and may not efficiently expose the epitopes or spatial configurations necessary for PLA probe binding and ligation. Consequently, tetrameric structures might not be effectively detected by α-synuclein-PLA. Furthermore, physiological tetramers are fragile during tissue processing, and their detection requires preservation of lipid-protein interactions, which are lost during standard histological procedures. In contrast, pathological oligomers are more stable, likely surviving such processing and being detectable by PLA.

Clinical implications and biomarker potential

Biochemically, α-synuclein oligomers have been detected in human postmortem brain extracts from patients with PD and DLB [243]. Significantly more synuclein oligomers were found in PD brains than in AD brains without Lewy-related pathology [429]. Furthermore, α-synuclein oligomers have also been detected in cerebrospinal fluid in PD and DLB, indicating their potential use as a biomarker for the underlying α-synuclein pathology [430, 448452]. Hansson et al. demonstrated that α-synuclein oligomer levels in cerebrospinal fluid were higher in PDD compared to normal controls and AD; in DLB they were not significantly different from those in controls but higher than in AD [453]. van Steenoven et al. demonstrated that a combination of tau and α-synuclein oligomers in cerebrospinal fluid could differentiate DLB and AD [454].

Regarding the clinical implication of α-synuclein oligomers, Majbour et al. revealed that patients with advanced disease (Hoehn & Yahr stage II or higher) had significantly elevated α-synuclein oligomer levels in cerebrospinal fluid. With longitudinal assessment, they demonstrated that oligomer values at the 48-month timepoint strongly correlated with motor symptom severity [451]. Furthermore, a recent study demonstrated that PDD had more abundant α-synuclein oligomers in the hippocampus compared to PD without cognitive impairment, while Lewy-related pathology was comparable between those with and without cognitive impairment [447]. An association between cognitive impairment and α-synuclein oligomers has also been suggested in MSA [440, 446]. Additionally, another recent study in DLB found that patients with rapid cognitive decline had more abundant α-synuclein oligomers in the CA1 subfield of the hippocampus compared to those with slow cognitive decline [455]. With this recently developed visualization technique, further studies on the clinicopathological correlates of α-synuclein oligomers are warranted.

Comorbid pathology – frequency and clinical impact

Aging is the major risk factor for most neurodegenerative diseases [456] and cerebrovascular disease [457]. Therefore, comorbid pathologies have been reported in neurodegenerative disorders [458463]. In this section, we summarize previous reports of comorbid pathology from various investigators and discuss the clinicopathological impact of each comorbid pathology. Figure 5 illustrates the frequencies of comorbid pathologies based on previous reports and data from our institution. The overall frequency of each comorbid pathology was estimated using a forest plot. When multiple reports were published from the same institution, the representative one was adopted.

Fig. 5.

Fig. 5

Frequency of each comorbid pathology. AD, Alzheimer’s disease; AGD, argyrophilic grain disease; CAA, cerebral amyloid angiopathy; CSVD, cerebral small vessel disease; TDP-43, transactive response DNA binding protein 43 kDa; (a) data from University of Kentucky Alzheimer’s disease Center; (b) data from national Alzheimer’s coordinating center Registry; (c) data from Tokyo institute of Psychiatry; (d) data from Canadian collaborative cohort of related dementia study

AD neuropathologic change

Among comorbid pathologies, AD neuropathologic change is the most commonly observed in Lewy body disease. The pathology of AD is characterized by a combination of neurofibrillary tangles (NFTs) and senile plaques. NFTs are composed of hyperphosphorylated tau, while senile plaques are made of amyloid β. The distribution of each pathology is assessed based on the Braak NFT stage [464] and Thal amyloid phase [465], respectively. Additionally, the density of neuritic plaques is assessed using the Consortium to Establish a Registry for Alzheimer’s Disease (CERAD) score [466]. AD neuropathologic change is taken into account to assess the pathologic findings in Lewy body disease [1] (Table 5). Population-based autopsy studies demonstrate that the prevalence of neuropathologically confirmed AD increases with age, with rates ranging from 12% to over 60% depending on age, cohort characteristics, and diagnostic criteria [83, 467469].

Table 5.

Association of pathology stage combinations with typical DLB syndrome likelihood

Alzheimer disease neuropathologic change (NIA-AA research framework)
Lewy-related pathology stage none/low intermediate high
Diffuse neocortical High High Intermediate
Limbic (transitional) High Intermediate Low
Brainstem-predominant Low Low Low
Amygdala-predominant Low Low Low
Olfactory bulb only Low Low Low

DLB, dementia with Lewy bodies; NIA-AA, National Institute on Aging and Alzheimer’s Association

With respect to the frequency of AD neuropathologic change in Lewy body dementia, in a community-based study conducted in the southern Rogaland County, Norway, 238 PD patients were identified and prospectively followed [268]. Of the 22 patients for whom autopsies were obtained, 18 had dementia, of which 15 had low, 3 had intermediate, and 0 had high probability of AD based on the National Institute on Aging (NIA)-Reagan Institute criteria [470]. Another study also reported that 13 out of 79 PDD patients had AD neuropathologic change [266]. Subsequent studies have reported varying frequencies of comorbid AD in PDD and DLB [4, 116, 259, 471475]. In the Lewy Body Dementia Center Without Walls (CWOW), Mayo Clinic brain bank, comorbid AD pathology was observed in 215 patients out of 363 Lewy body dementia patients (59%). We employed the Mayo Clinic Florida NIA-Alzheimer’s Association (NIA-AA) operational criteria to define AD neuropathologic change as previously described [463, 476]. Table 6 summarizes the frequency of comorbid AD and definition of AD employed in each study. Using these data, a forest plot estimated the frequency of AD complications in Lewy body dementia at 37% (Fig. 5 and Supplementary Fig. S1).

Table 6.

Frequency of comorbid Alzheimer’s disease pathology in Lewy body dementia

Author (year) N of Lewy body dementia cases N (%) of cases with AD Facility/Region Country Definition of AD
Aarsland et al. (2005) [268] 18 3 (17%) County of Rogaland Norway NIA-RI criteria [477]
Braak et al. (2005) [266] 79 13 (16%) Medisch Spectrum Twente Hospital Group in Enschede Netherlands Braak staging [464]
Galvin et al. (2006) [259] 34 10 (29%) Washington University Movement Disorder Center USA NIA-RI criteria [477]
Hely et al. (2008) [4] 17 3 (18%) Multicenter in Sydney Australia NIA-RI criteria [477]
Jellinger et al. (2008) [471] 20 5 (25%) Vienna Neurobiology brain bank Austria Braak staging [464]
Nelson et al. (2009) [472] 33 19 (58%) University of Kentucky Alzheimer’s Disease Center USA NIA-RI criteria [477]
Nelson et al. (2009) [472] 374 175 (47%) National Alzheimer’s Coordinating Center Registry data USA NIA-RI criteria [477]
Kotzbauer et al. (2012) [473] 32 1 (3%) Movement Disorders Center at Washington University School of Medicine USA NIA-RI criteria [477]
Colom-Cadena et al. (2017) [474] 47 29 (62%) Neurological Tissue Bank of the Biobanc-Hospital Clinic-IDIBAPS in Barcelona Spain NIA-AA guidelines [466]
Irwin et al. (2017) [116] 213 108 (51%) Five academic institutions USA NIA-AA guidelines [466]
McAleese et al. (2017) [475] 34 19 (56%) Newcastle Brain Tissue Resource UK NIA-AA guidelines [466]
Our data 363 215 (59%) Lewy Body Dementia Center Without Walls, Mayo Clinic brain bank USA Mayo Clinic Florida NIA-AA operational criteria [463, 476]

AD, Alzheimer’s disease; NIA-RI, National Institute on Aging–Reagan Institute; NIA-AA, National Institute on Aging and Alzheimer’s Association

Penn Udall Center for Excellence in Parkinson’s Disease Research (Philadelphia), Pacific Udall Center (Seattle and Portland), Alzheimer’s Disease Core Center (Philadelphia), Alzheimer’s Disease Research Center (Seattle), Layton Aging and Alzheimer’s Disease Center (Portland), Alzheimer’s Disease Research Center (Pittsburgh), and Sanders-Brown Center on Aging (Lexington)

To date, many studies have demonstrated direct molecular interactions of α-synuclein with amyloid β and tau. Co-incubation experiments have demonstrated that α-synuclein monomers and oligomers inhibit amyloid-β fibrillization while promoting toxic oligomer formation via the C-terminus of amyloid-β. [478, 479]. This suggests synergistic toxicity mechanisms between α-synuclein and amyloid-β. Another experiment shows that oligomeric forms of amyloid-β can trigger α-synuclein aggregation via heterogeneous primary nucleation [480]. Additionally, amyloid-β and α-synuclein have a cross-seeding effect in vitro with fibrils showing stronger seeding activity than oligomers [481]. Similarly, α-synuclein and tau exhibit bidirectional interactions. Direct binding between α-synuclein and tau has been demonstrated, with the C-terminus of α-synuclein selectively interacting with tau and promoting its aggregation [482485]. Additionally, α-synuclein and tau have been shown to synergistically promote each other’s aggregation [482, 483, 486490]. The C-terminal region of α-synuclein plays an important role and α-synuclein phosphorylation at Ser129 further enhances tau aggregation [483, 487]. These studies collectively provide experimental evidence for direct protein-protein interactions, cross-aggregation, and synergistic effects between α-synuclein and both amyloid-β and tau, supporting mechanistic links underlying their co-occurrence.

The clinicopathological impact of comorbid AD pathology has been studied. With respect to the impact on clinical diagnosis, when AD pathology is comorbid with Lewy body disease, the probability of accurate antemortem diagnosis decreases, and patients frequently receive a clinical diagnosis of AD alone [491]. Regarding the impact on clinical manifestations, in a study comparing PDD with and without AD, PDD with AD had a higher age at PD onset and at death, shorter time to onset of dementia, and shorter duration of PD symptoms before dementia onset [492]. Another study found more severe tau and amyloid pathology in addition to Lewy-related pathology in PDD compared to PD without dementia [493]. PD with and without AD pathology exhibited a different distribution of Lewy-related pathology and PD with AD pathology was more likely to develop dementia compared to PD without AD pathology [494].

In contrast, several studies have reported that AD comorbidity does not have a major impact on cognitive function. One comparative study of neuropsychological assessment reported that although visuospatial impairment was more severe in DLB with AD than DLB without AD, cognitive decline itself was not significantly different between DLB with or without AD [495]. In another study, while Boston Naming Test was worse in Lewy body dementia with AD, MMSE, Dementia Rating Scale, and category fluency were comparable in those with and without AD [496]. Furthermore, a recent study examined the effect of 11 pathological conditions, including AD and Lewy-related pathology, on cognitive decline and demonstrated that these pathologies could not fully explain the progression of cognitive decline [497]. Another recent study compared the rate of MMSE decline among pure AD, AD with diffuse Lewy body disease, and AD with mild Lewy body pathology (to the extent that diffuse distribution and density thresholds were not met) and found that AD with mild Lewy body pathology had the fastest MMSE decline rate [498]. Although it is difficult to explain why AD with mild Lewy body pathology has a faster MMSE decline than AD+diffuse Lewy body disease, the authors speculated that cortical Lewy bodies may have some protective mechanism against AD pathology. As has been reviewed, the impact of comorbid AD pathology on cognitive function in Lewy body dementia remains controversial and is not fully understood.

TAR DNA-binding protein 43 (TDP-43) pathology

TAR DNA-binding protein 43 (TDP-43) is a ubiquitously expressed nuclear protein and plays an important role in essential cell functions, including regulation of RNA metabolism, RNA splicing, mRNA transport, microRNA maturation, translational regulation, and the formation of stress granules [499]. Pathological aggregates of TDP-43 have been identified in the cytoplasm of neurons and glial cells in brains of patients with amyotrophic lateral sclerosis and frontotemporal lobar degeneration [500, 501]. According to population-based autopsy studies, the prevalence of TDP-43 pathology in the general population ranges from 19% to 64% [502505].

Comorbid TDP-43 pathology has been reported in neurodegenerative disorders including Lewy body disease and AD [460, 474, 475, 506509]. Table 7 summarizes the frequency of comorbid TDP-43 pathology in Lewy body dementia in previous studies. In the Lewy Body Dementia CWOW, Mayo Clinic brain bank, comorbid TDP-43 pathology was observed in 119 out of 350 patients with Lewy body dementia (34%). Based on these data, the overall frequency of TDP-43 pathology in Lewy body dementia was 36% (Fig. 5 and Supplementary Fig. S2).

Table 7.

Frequency of comorbid TDP-43 pathology in Lewy body dementia

Author (year) N of Lewy body dementia cases N (%) of cases with TDP-43 Facility/Region Country Definition of TDP-43
Nakashima-Yasuda et al. (2007) [506] 111 29 (26%) Five academic institutions USA Presence of TDP-43-positive inclusions on IHC with antibody against TDP-43 (rabbit, polyclonal, ProteinTech)
Higashi et al. (2007) [507] 11 5 (45%) Juntendo University Japan Presence of TDP-43-positive inclusions on IHC with antibody against TDP-43 (rabbit, polyclonal, ProteinTech)
Arai et al. (2009) [508] 15 8 (53%) Tokyo Institute of Psychiatry Japan Presence of TDP-43-positive inclusions on IHC with antibody against phosphorylated TDP-43 (pS409/410 and pS403/404) [510]
Arai et al. (2009) [508] 10 6 (60%) Canadian Collaborative Cohort of Related Dementia study Canada Presence of TDP-43-positive inclusions on IHC with antibody against phosphorylated TDP-43 (pS409/410 and pS403/404) [510]
Colom-Cadena et al. (2017) [474] 47 10 (21%) Neurological Tissue Bank of the Biobanc-Hospital Clinic-IDIBAPS in Barcelona Spain Presence of TDP-43-positive inclusions on IHC with antibody against TDP-43 (2E2–D3, mouse, monoclonal, Abnova)
McAleese et al. (2017) [475] 34 15 (44%) Newcastle Brain Tissue Resource UK Presence of TDP-43-positive inclusions on IHC with antibody against phosphorylated TDP-43 (pS409/410–2, rabbit, polyclonal, Cosmo Bio)
Our data 350 119 (34%) Lewy Body Dementia Center Without Walls, Mayo Clinic brain bank USA Presence of TDP-43-positive inclusions on IHC with antibody against phosphorylated TDP-43 (pS409/410, mouse monoclonal, Cosmo Bio) [511]

TDP-43, transactive response DNA binding protein 43 kDa

† Penn Udall Center for Excellence in Parkinson’s Disease Research (Philadelphia), Pacific Udall Center (Seattle and Portland), Alzheimer’s Disease Core Center (Philadelphia), Alzheimer’s Disease Research Center (Seattle), Layton Aging and Alzheimer’s Disease Center (Portland), Alzheimer’s Disease Research Center (Pittsburgh), and Sanders-Brown Center on Aging (Lexington)

Several studies have investigated molecular interactions between α-synuclein and TDP-43. One study demonstrated that the low complexity domain (amino acids 310–329) of TDP-43 binds to full-length α-synuclein, while the non-amyloid β component of α-synuclein binds to full-length TDP-43 [512]. Additionally, the prion-like pathogenic C-terminal domain of TDP-43 interacts with α-synuclein to generate hybrid fibrils [513, 514]. Interestingly, α-synuclein monomers, oligomers, and sonicated fibrils seeded TDP-43 prion-like pathogenic C-terminal domain monomers, whereas reverse seeding did not occur. This suggests that TDP-43 abnormal aggregation may be secondarily induced by α-synuclein aggregation.

Regarding the effect on clinical diagnosis, patients with hippocampal TDP-43 were less likely diagnosed with DLB during their life and often received an alternative diagnosis of AD [515]. For the impact on clinical manifestations, whereas TDP-43 pathology deteriorates cognitive function in AD [516], its impact in Lewy body dementia appears to be limited. One study revealed that while disease duration was not significantly different, age at death was higher in PDD with TDP-43 compared to PDD without TDP-43 [506]. Another study did not find significant differences in age at death, disease duration, NFT stage, and Lewy body stage between DLB with and without TDP-43 [507]. No significant differences were observed in age at onset of cognitive impairment, cognitive decline progression, and final MMSE score between DLB with and without TDP-43 pathology [475]. However, a recent study with larger number of patients demonstrated that Lewy body disease with limbic-predominant TDP-43 had lower MMSE and faster cognitive decline compared to those without TDP-43 [517]. Thus, TDP-43 may independently exacerbate cognitive function in Lewy body dementia, and further studies are warranted.

Cerebral small vessel disease (CSVD)

CSVD encompasses a range of pathological changes affecting the small arteries, arterioles, capillaries, and venules in the brain and can cause dementia, i.e., vascular dementia [518520]. Pathologically, CSVD is characterized by several features: cortical and subcortical infarction, hemorrhage, microinfarcts, microbleeds, white matter change, and cerebral amyloid angiopathy (CAA). Among them, CAA will be discussed separately in the next section. As the focus of this review is pathology, MRI studies are not discussed.

Population-based autopsy studies show CSVD prevalence of 19% to 39% in the general population [468, 469]. Frequency of comorbid CSVD in Lewy body dementia ranged from 16% to 68% (Table 8) [4, 258, 471, 521, 523]. In the Lewy Body Dementia CWOW, Mayo Clinic brain bank, comorbid CSVD was detected in 84 out of 363 patients with Lewy body dementia (23%). Based on these data, the overall frequency of CSVD in Lewy body dementia was 33% (Fig. 5 and Supplementary Fig. S3).

Table 8.

Frequency of comorbid cerebral small vessel disease in Lewy body dementia

Author (year) N of Lewy body dementia cases N (%) of cases with CSVD Facility/Region Country Definition of CSVD
Isojima et al. (2006) [521] 25 17 (68%) Yokohama City University and Fukushimura Hospital Japan Presence of hemorrhage, infarcts, or microinfarcts
Hely et al. (2008) [4] 17 5 (29%) Multicenter in Sydney Australia NA
Jellinger et al. (2008) [471] 66 22 (33%) Vienna Neurobiology brain bank Austria Presence of lacunes, infarcts, microinfarcts, hemorrhage, white matter lesions, or subcortical arteriosclerotic encephalopathy [522]
Ghebremedhin et al. (2010) [523] 13 5 (38%) Medisch Spectrum Twente Hospital Group in Enschede Netherlands Presence of arteriosclerotic, arteriolosclerotic, lipohyalinotic (fibrinoid necrosis), or microaneurysmal formation in small parenchymal brain vessels [524]
Horvath et al. (2013) [258] 109 17 (16%) Geneva University Brain Bank Switzerland Presence of infarcts, microinfarcts, or lacunar state
Our data 363 84 (23%) Lewy Body Dementia Center Without Walls, Mayo Clinic brain bank USA Presence of arteriolosclerosis with microinfarcts, microbleeds, or ischemic white matter changes [463]

CSVD, cerebral small vessel disease

In a study examining the correlation between the presence of CSVD and clinical symptoms in DLB, memory disturbance was more frequent in DLB with CSVD, and parkinsonism was more frequent in DLB with CSVD as initial manifestations, whereas there was no difference in the frequency of individual clinical symptoms over the entire clinical course [521]. In a recent study using National Alzheimer’s Coordinating Center Registry data, hemorrhage and microbleeds had weak positive correlation with digit span forward length, but not with memory or language domains, while infarcts, lacunar, and microinfarcts did not correlate with either battery [525]. Pathologically, an inverse association between Lewy body score and small vessel disease score has been reported, and the authors have proposed that CSVD may lower the threshold for symptom development [523]. Besides these findings, there are reports that CSVD is associated with parkinsonism [526] and autonomic dysfunction [527]; however, its impact on cognitive function in Lewy body dementia seems limited.

Cerebral amyloid angiopathy (CAA)

CAA is pathologically defined by the amyloid β deposition in the walls of small and medium-sized blood vessels in the cerebral cortex and leptomeninges [528]. The prevalence of CAA in the general population ranges from 40% to 79% according to population-based autopsy findings [529532]. In previous autopsy studies, the frequency of comorbid CAA ranged from 18% to 82% in Lewy body dementia (Table 9) [4, 72, 266, 474, 475, 525]. In the Lewy Body Dementia CWOW, Mayo Clinic brain bank, 131 out of 363 patients exhibited comorbid CAA (36%). The overall frequency of comorbid CAA was estimated at 49% (Fig. 5 and Supplementary Fig. S4). One study including 110 PDD and 60 DLB demonstrated that the frequency of comorbid CAA was higher in DLB than in PDD (90% vs. 50%) [72].

Table 9.

Frequency of comorbid cerebral amyloid angiopathy in Lewy body dementia

Author (year) N of Lewy body dementia cases N (%) of cases with CAA Facility/Region Country Definition of CAA
Braak et al. (2005) [266] 79 29 (37%) Medisch Spectrum Twente Hospital Group in Enschede Netherlands Revesz et al. [533]
Hely et al. (2008) [4] 17 3 (18%) Multicenter in Sydney Australia NA
Colom-Cadena et al. (2017) [474] 47 26 (55%) Neurological Tissue Bank of the Biobanc-Hospital Clinic-IDIBAPS in Barcelona Spain Vonsattel et al. [534]
McAleese et al. (2017) [475] 34 28 (82%) Newcastle Brain Tissue Resource UK Thal et al. [535]
Jellinger (2021) [72] 170 109 (64%) Vienna Neurobiology brain bank Austria Love et al. [536]
Chen et al. (2022) [525] 363 131 (36%) National Alzheimer’s Coordinating Center Registry data USA Vonsattel et al. [534]
Our data 360 164 (46%) Lewy Body Dementia Center Without Walls, Mayo Clinic brain bank USA Presence of amyloid deposition on vessel walls by thioflavin-S fluorescent microscopy [463]

CAA, cerebral amyloid angiopathy

The severity of CAA has been associated with a rapid disease progression in DLB compared to PDD [72]. A recent study using National Alzheimer’s Coordinating Center Registry data demonstrated a negative correlation between comorbid CAA and logical memory in the intermediate likelihood DLB [525]. This correlation was not observed in the high likelihood DLB. A few studies have examined the clinical impact of comorbid CAA, and further research is needed.

Argyrophilic grain disease (AGD)

AGD is a distinct tauopathy characterized by the presence of spindle- or comma-shaped tau aggregates (argyrophilic grains) [537, 538]. Clinically, AGD can present with cognitive impairment [539, 540]. Although the prevalence of AGD in the general population remains unclear from population-based autopsy studies, consecutive autopsy series report AGD prevalence rates of 5–9% [541543]. In Lewy body dementia, the frequency of comorbid AGD ranged from 8% to 83% in autopsy studies (Table 10) [266, 474, 515, 546, 547]. In a study examining 367 autopsy cases, AGD was found in 18% of AD, 26% of PDD, 22% of DLB, and 29% of normal controls, with no significant differences in frequency among conditions. [546]. The overall frequency of comorbid AGD in Lewy body dementia is estimated at 34% (Fig. 5 and Supplementary Fig. S5). Whether comorbid AGD independently affects clinical manifestations, including cognitive decline and Lewy-related pathology, remains unstudied.

Table 10.

Frequency of comorbid argyrophilic grain disease in Lewy body dementia

Author (year) N of Lewy body dementia cases N (%) of cases with AGD Facility/Region Country Definition of AGD
Braak et al. (2005) [266] 79 29 (37%) Medisch Spectrum Twente Hospital Group in Enschede Netherlands Braak et al. [544], Jellinger [537], and Iseki et al. [545]
Sabbagh et al. (2009) [546] 51 13 (25%) Sun Health Research Institute Brain Donation Program USA Presence of grains on Gallyas silver staining
Homma et al. (2015) [547] 12 10 (83%) Tokyo Metropolitan Neurological Hospital Japan Presence of argyrophilic grains on Gallyas–Braak staining and Saito staging [548]
Colom-Cadena et al. (2017) [474] 47 10 (21%) Neurological Tissue Bank of the Biobanc-Hospital Clinic-IDIBAPS in Barcelona Spain Saito staging [548]
Buciuc et al. (2020) [515] 158 12 (8%) Mayo Clinic Alzheimer’s Disease Research Center USA Presence of silver and tau-positive spindle-shaped lesions [537]

AGD, argyrophilic grain disease

Conclusions

This review highlights the complex neuropathological landscape of Lewy body dementia, encompassing classical Lewy-related pathology, α-synuclein oligomers, and comorbid pathologies along with their clinical implications. While significant progress has been made in understanding the underlying pathology of the disease, many questions remain unanswered. Specifically, the origin of pathological α-synuclein, the detailed pathways of its propagation, and the clinical impact of α-synuclein oligomers and each comorbid pathology are still largely unresolved. Future research should focus on elucidating the origins of pathological α-synuclein, pathways of its propagation, as well as the interplay between α-synuclein and comorbid pathologies. Such efforts are essential to advancing our knowledge and improving clinical outcomes for individuals affected by Lewy body dementia.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Acknowledgements

Not applicable.

Abbreviations

AD

Alzheimer’s disease

AGD

Argyrophilic grain disease

CAA

Cerebral amyloid angiopathy

CERAD

Consortium to establish a registry for Alzheimer’s Disease

CSVD

Cerebral small vessel disease

CWOW

Center without walls

DLB

Dementia with Lewy bodies

DSM-5

Diagnostic and statistical manual of mental disorders, Fifth Edition

MMSE

Mini-Mental state examination

MSA

Multiple system atrophy

NFT

Neurofibrillary tangle

NIA

National institute on aging

NIA-AA

National institute on aging-Alzheimer’s association

PD

Parkinson’s disease

PDD

Parkinson’s disease dementia

PLA

Proximity ligation assay

RBD

Rapid eye movement sleep behavior disorder

SuStaIn

Subtype and stage inference

TDP-43

TAR DNA-binding protein 43

Author contributions

H.S., T.M, and D.W.D, contributed to the conception and design of the study; H.S. and T.M. contributed to the preparation of the figures; H.S., T.M., M.A.D, and D.W.D. contributed to drafting/revision of the text; all authors have read and agreed to the final manuscript.

Funding

Not applicable.

Data availability

Not applicable.

Declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that there are no competing interests.

Footnotes

Publisher’s Note

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

Contributor Information

Hiroaki Sekiya, Email: sekiya.hiroaki@mayo.edu.

Dennis W. Dickson, Email: dickson.dennis@mayo.edu

References

  • 1.McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor JP, Weintraub D, Aarsland D, Galvin J, Attems J, Ballard CG, et al. Diagnosis and management of dementia with Lewy bodies: fourth consensus report of the DLB consortium. Neurology. 2017;89:88–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Poewe W. Non-motor symptoms in Parkinson’s disease. Eur J Neurol. 2008;15(Suppl, 1):14–20. [DOI] [PubMed] [Google Scholar]
  • 3.Svenningsson P, Westman E, Ballard C, Aarsland D. Cognitive impairment in patients with Parkinson’s disease: diagnosis, biomarkers, and treatment. The Lancet Neurol. 2012;11:697–707. [DOI] [PubMed] [Google Scholar]
  • 4.Hely MA, Reid WG, Adena MA, Halliday GM, Morris JG. The Sydney multicenter study of Parkinson’s disease: the inevitability of dementia at 20 years. Mov Disord. 2008;23:837–44. [DOI] [PubMed] [Google Scholar]
  • 5.Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–40. [DOI] [PubMed] [Google Scholar]
  • 6.Baba M, Nakajo S, Tu PH, Tomita T, Nakaya K, Lee VM, Trojanowski JQ, Iwatsubo T. Aggregation of alpha-synuclein in Lewy bodies of sporadic Parkinson’s disease and dementia with Lewy bodies. Am J Pathol. 1998;152:879–84. [PMC free article] [PubMed] [Google Scholar]
  • 7.Nors Perdersen M, V F, Horvath I, van Maarschalkerweerd A, Nørgaard Toft, Weise C, Almqvist F, Wolf-Watz M, Wittung-Stafshede P, Vestergaard B. Direct correlation between ligand-induced α-Synuclein oligomers and amyloid-like fibril growth. Sci Rep. 2015;5:10422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Alam P, Bousset L, Melki R, Otzen DE. α-synuclein oligomers and fibrils: a spectrum of species, a spectrum of toxicities. J Neurochem. 2019;150:522–34. [DOI] [PubMed] [Google Scholar]
  • 9.Ingelsson M. Alpha-Synuclein oligomers-neurotoxic molecules in Parkinson’s disease and other Lewy body disorders. Front Neurosci. 2016;10:408. [DOI] [PMC free article] [PubMed]
  • 10.Bengoa-Vergniory N, Roberts RF, Wade-Martins R, Alegre-Abarrategui J. Alpha-synuclein oligomers: a new hope. Acta Neuropathol. 2017;134:819–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bigi A, Cascella R, Cecchi C. α-synuclein oligomers and fibrils: partners in crime in synucleinopathies. Neural Regen Res. 2023;18:2332–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Smith C, Malek N, Grosset K, Cullen B, Gentleman S, Grosset DG. Neuropathology of dementia in patients with Parkinson’s disease: a systematic review of autopsy studies. J Neurol Neurosurg Psychiatry. 2019;90:1234–43. [DOI] [PubMed] [Google Scholar]
  • 13.Schneider JA. Neuropathology of dementia disorders. Continuum (minneap Minn). 2022;28:834–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Toledo JB, Abdelnour C, Weil RS, Ferreira D, Rodriguez-Porcel F, Pilotto A, Wyman-Chick KA, Grothe MJ, Kane JPM, Taylor A, et al. Dementia with Lewy bodies: impact of co-pathologies and implications for clinical trial design. Alzheimers Dement. 2023;19:318–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Petersen RC, Weintraub S, Sabbagh M, Karlawish J, Adler CH, Dilworth-Anderson P, Frank L, Huling Hummel C, Taylor A. A new framework for dementia nomenclature. JAMA Neurol. 2023;80:1364–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Association AP. Diagnostic and statistical manual of mental disorders: DSM-5. American Psychi Assoc. 2013. [Google Scholar]
  • 17.Goedert M, Jakes R, Spillantini MG. The synucleinopathies: twenty years on. J Parkinsons Dis. 2017;7:S51–s69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Dickson DW, Lin W, Liu WK, Yen SH. Multiple system atrophy: a sporadic synucleinopathy. Brain Pathol. 1999;9:721–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Trojanowski JQ. Revesz T. Proposed neuropathological criteria for the post mortem diagnosis of multiple system atrophy. Neuropathol Appl Neurobiol. 2007;33:615–20. [DOI] [PubMed] [Google Scholar]
  • 20.Papp MI, Kahn JE, Lantos PL. Glial cytoplasmic inclusions in the CNS of patients with multiple system atrophy (striatonigral degeneration, olivopontocerebellar atrophy and shy-drager syndrome). J Neurol Sci. 1989;94:79–100. [DOI] [PubMed] [Google Scholar]
  • 21.Cykowski MD, Coon EA, Powell SZ, Jenkins SM, Benarroch EE, Low PA, Schmeichel AM, Parisi JE. Expanding the spectrum of neuronal pathology in multiple system atrophy. Brain. 2015;138:2293–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sekiya H, Kowa H, Koga H, Takata M, Satake W, Futamura N, Funakawa I, Jinnai K, Takahashi M, Kondo T, et al. Wide distribution of alpha-synuclein oligomers in multiple system atrophy brain detected by proximity ligation. Acta Neuropathol. 2019;137:455–66. [DOI] [PubMed] [Google Scholar]
  • 23.Wiseman JA, Halliday GM, Dieriks BV. Neuronal α-synuclein toxicity is the key driver of neurodegeneration in multiple system atrophy. Brain. 2025. [DOI] [PMC free article] [PubMed]
  • 24.Aarsland D, Zaccai J, Brayne C. A systematic review of prevalence studies of dementia in Parkinson’s disease. Mov Disord. 2005;20:1255–63. [DOI] [PubMed] [Google Scholar]
  • 25.Planas-Ballvé A, Rios J, Ispierto L, Gea M, Grau L, Jiménez M, Cáceres C, Martínez S, Beyer K, Álvarez R, et al. Key motor and non-motor features in early dementia with Lewy bodies. Front Neurol. 2025;16:1555175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Choudhury P, Zhang N, Adler CH, Chen K, Belden C, Driver-Dunckley E, Mehta SH, Shprecher DR, Serrano GE, Shill HA, et al. Longitudinal motor decline in dementia with Lewy bodies, Parkinson disease dementia, and Alzheimer’s dementia in a community autopsy cohort. Alzheimers Dement. 2023;19:4377–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Onofrj M, Varanese S, Bonanni L, Taylor JP, Antonini A, Valente EM, Petrucci S, Stocchi F, Thomas A, Perfetti B. Cohort study of prevalence and phenomenology of tremor in dementia with Lewy bodies. J Neurol. 2013;260:1731–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Auning E, Rongve A, Fladby T, Booij J, Hortobágyi T, Siepel FJ, Ballard C, Aarsland D. Early and presenting symptoms of dementia with Lewy bodies. Dement Geriatr Cogn Disord. 2011;32:202–08. [DOI] [PubMed] [Google Scholar]
  • 29.Rana AQ, Vaid HM, Edun A, Dogu O, Rana MA. Relationship of dementia and visual hallucinations in tremor and non-tremor dominant Parkinson’s disease. J Neurol Sci. 2012;323:158–61. [DOI] [PubMed] [Google Scholar]
  • 30.Baumann CR, Held U, Valko PO, Wienecke M, Waldvogel D. Body side and predominant motor features at the onset of Parkinson’s disease are linked to motor and nonmotor progression. Mov Disord. 2014;29:207–13. [DOI] [PubMed] [Google Scholar]
  • 31.van der Heeden Jf, Marinus J, Martinez-Martin P, Rodriguez-Blazquez C, Geraedts VJ, van Hilten Jj. Postural instability and gait are associated with severity and prognosis of Parkinson disease. Neurology. 2016;86:2243–50. [DOI] [PubMed] [Google Scholar]
  • 32.Mayo AM, Peavy GM. Associations among Braak stage, parkinsonian gait, cognition, and functional status in autopsy-confirmed dementia with Lewy bodies. Int J Geriatr Psychiatry. 2019;34:738–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Müller ML, Frey KA, Petrou M, Kotagal V, Koeppe RA, Albin RL, Bohnen NI. β-amyloid and postural instability and gait difficulty in Parkinson’s disease at risk for dementia. Mov Disord. 2013;28:296–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Allan LM, Ballard CG, Rowan EN, Kenny RA. Incidence and prediction of falls in dementia: a prospective study in older people. PLoS One. 2009;4:e 5521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Cheng KL, Lin LH, Chen PC, Chiang PL, Chen YS, Chen HL, Chen MH, Chou KH, Li SH, Lu CH, Lin WC. Reduced gray matter volume and risk of falls in Parkinson’s disease with dementia patients: a voxel-based morphometry study. Int J Environ Res Public Health. 2020;17. [DOI] [PMC free article] [PubMed]
  • 36.Nedelec T, Couvy-Duchesne B, Darves-Bornoz A, Couronne R, Monnet F, Gantzer L, Lekens B, Wu Y, Villain N, Schrag A, et al. A comparison between early presentation of dementia with Lewy Bodies, Alzheimer’s disease, and Parkinson’s disease: evidence from routine primary care and UK Biobank Data. Ann Neurol. 2023;94:259–70. [DOI] [PubMed] [Google Scholar]
  • 37.Howell MJ, Schenck CH. Rapid eye movement sleep behavior disorder and neurodegenerative disease. JAMA Neurol. 2015;72:707–12. [DOI] [PubMed] [Google Scholar]
  • 38.St Louis EK, Boeve AR, Boeve BF. REM sleep behavior disorder in Parkinson’s disease and other synucleinopathies. Mov Disord. 2017;32:645–58. [DOI] [PubMed] [Google Scholar]
  • 39.Fernández-Arcos A, Morenas-Rodríguez E, Santamaria J, Sánchez-Valle R, Lladó A, Gaig C, Lleó A, Iranzo A. Clinical and video-polysomnographic analysis of rapid eye movement sleep behavior disorder and other sleep disturbances in dementia with Lewy bodies. Sleep. 2019;42. [DOI] [PubMed]
  • 40.Hu MT. REM sleep behavior disorder (RBD). Neurobiol Dis. 2020;143:104996. [DOI] [PubMed] [Google Scholar]
  • 41.Stang CD, Mullan AF, Hajeb M, Camerucci E, Turcano P, Martin P, Mielke MM, Josephs KA, Bower JH, St Louis EK, et al. Timeline of rapid eye movement Sleep behavior disorder in overt alpha-synucleinopathies. Ann Neurol. 2021;89:293–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Choudhury P, Graff-Radford J, Aakre JA, Wurtz L, Knopman DS, Graff-Radford NR, Kantarci K, Forsberg LK, Fields JA, Pedraza O, et al. The temporal onset of the core features in dementia with Lewy bodies. Alzheimers Dement. 2022;18:591–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Pagano G, De Micco R, Yousaf T, Wilson H, Chandra A, Politis M. REM behavior disorder predicts motor progression and cognitive decline in Parkinson disease. Neurology. 2018;91:e894–905. [DOI] [PubMed] [Google Scholar]
  • 44.Elder GJ, Lazar AS, Alfonso-Miller P, Taylor JP. Sleep disturbances in Lewy body dementia: a systematic review. Int J Geriatr Psychiatry. 2022;37. [DOI] [PMC free article] [PubMed]
  • 45.Baumann-Vogel H, Hor H, Poryazova R, Valko P, Werth E, Baumann CR. REM sleep behavior in Parkinson disease: frequent, particularly with higher age. PLoS One. 2020;15:e0243454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Mosimann UP, Rowan EN, Partington CE, Collerton D, Littlewood E, O’Brien JT, Burn DJ, McKeith IG. Characteristics of visual hallucinations in Parkinson disease dementia and dementia with Lewy bodies. Am J Geriatr Psychiatry. 2006;14:153–60. [DOI] [PubMed] [Google Scholar]
  • 47.Archibald NK, Clarke MP, Mosimann UP, Burn DJ. Visual symptoms in Parkinson’s disease and Parkinson’s disease dementia. Mov Disord. 2011;26:2387–95. [DOI] [PubMed] [Google Scholar]
  • 48.Lin W, Xie YC, Cheng PY, Dong LY, Hung GU, Chiu PY. Association of visual hallucinations with very mild degenerative dementia due to dementia with Lewy bodies. PLoS One. 2018;13:e0205909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Aarsland D, Ballard C, Larsen JP, McKeith I. A comparative study of psychiatric symptoms in dementia with Lewy bodies and Parkinson’s disease with and without dementia. Int J Geriatr Psychiatry. 2001;16:528–36. [DOI] [PubMed] [Google Scholar]
  • 50.Savica R, Grossardt BR, Bower JH, Boeve BF, Ahlskog JE, Rocca WA. Incidence of dementia with Lewy bodies and Parkinson disease dementia. JAMA Neurol. 2013;70:1396–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chiu PY, Tsai CT, Chen PK, Chen WJ, Lai TJ. Neuropsychiatric symptoms in Parkinson’s disease dementia are more similar to Alzheimer’s disease than dementia with Lewy bodies: a case-control study. PLoS One. 2016;11:e0153989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Eversfield CL, Orton LD. Auditory and visual hallucination prevalence in Parkinson’s disease and dementia with Lewy bodies: a systematic review and meta-analysis. Psychol Med. 2019;49:2342–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Ballard CG, Aarsland D, McKeith I, O’Brien J, Gray A, Cormack F, Burn D, Cassidy T, Starfeldt R, Larsen JP, et al. Fluctuations in attention: PD dementia vs DLB with parkinsonism. Neurology. 2002;59:1714–20. [DOI] [PubMed] [Google Scholar]
  • 54.Smirnov DS, Galasko D, Edland SD, Filoteo JV, Hansen LA, Salmon DP. Cognitive decline profiles differ in Parkinson disease dementia and dementia with Lewy bodies. Neurology. 2020 2087;94:e2076–e. [DOI] [PMC free article] [PubMed]
  • 55.Palermo G, Belli E, Tommasini L, Morganti R, Frosini D, Nicoletti V, Tognoni G, Siciliano G, Bonuccelli U, Baldacci F, Ceravolo R. Dissecting the interplay between time of dementia and cognitive profiles in Lewy Body Dementias. J Alzheimers Dis. 2021;84:757–66. [DOI] [PubMed] [Google Scholar]
  • 56.Allan LM, Ballard CG, Allen J, Murray A, Davidson AW, McKeith IG, Kenny RA. Autonomic dysfunction in dementia. J Neurol Neurosurg Psychiatry. 2007;78:671–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Stubendorff K, Aarsland D, Minthon L, Londos E. The impact of autonomic dysfunction on survival in patients with dementia with Lewy bodies and Parkinson’s disease with dementia. PLoS One. 2012;7:e45451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Horimoto Y, Matsumoto M, Akatsu H, Ikari H, Kojima K, Yamamoto T, Otsuka Y, Ojika K, Ueda R, Kosaka K. Autonomic dysfunctions in dementia with Lewy bodies. J Neurol. 2003;250:530–33. [DOI] [PubMed] [Google Scholar]
  • 59.Ransmayr GN, Holliger S, Schletterer K, Heidler H, Deibl M, Poewe W, Madersbacher H, Kiss G. Lower urinary tract symptoms in dementia with Lewy bodies, Parkinson disease, and Alzheimer disease. Neurology. 2008;70:299–303. [DOI] [PubMed] [Google Scholar]
  • 60.Tateno F, Sakakibara R, Ogata T, Kishi M, Tsuyusaki Y, Takahashi O, Sugiyama M, Tateno A. Lower urinary tract function in dementia with Lewy bodies (DLB). Mov Disord. 2015;30:411–15. [DOI] [PubMed] [Google Scholar]
  • 61.Li FF, Cui YS, Yan R, Cao SS. Feng T. Prevalence of lower urinary tract symptoms, urinary incontinence and retention in Parkinson’s disease: a systematic review and meta-analysis. Front Aging Neurosci. 2022;14:977572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Harding AJ, Halliday GM. Cortical Lewy body pathology in the diagnosis of dementia. Acta Neuropathol. 2001;102:355–63. [DOI] [PubMed] [Google Scholar]
  • 63.Tsuboi Y, Dickson DW. Dementia with Lewy bodies and Parkinson’s disease with dementia: are they different? Parkinsonism Relat Disord. 2005;11(Suppl, 1):S47–51. [DOI] [PubMed] [Google Scholar]
  • 64.Tsuboi Y, Uchikado H, Dickson DW. Neuropathology of Parkinson’s disease dementia and dementia with Lewy bodies with reference to striatal pathology. Parkinsonism Relat Disord. 2007;13(Suppl, 3):S221–224. [DOI] [PubMed] [Google Scholar]
  • 65.Kalaitzakis ME, Pearce RK, Gentleman SM. Clinical correlates of pathology in the claustrum in Parkinson’s disease and dementia with Lewy bodies. Neurosci Lett. 2009;461:12–15. [DOI] [PubMed] [Google Scholar]
  • 66.Kalaitzakis ME, Walls AJ, Pearce RK, Gentleman SM. Striatal Aβ peptide deposition mirrors dementia and differentiates DLB and PDD from other parkinsonian syndromes. Neurobiol Dis. 2011;41:377–84. [DOI] [PubMed] [Google Scholar]
  • 67.Sierra M, Gelpi E, Martí MJ. Compta Y. Lewy- and alzheimer-type pathologies in midbrain and cerebellum across the Lewy body disorders spectrum. Neuropathol Appl neurobiol. 2016;42:451–62. [DOI] [PubMed] [Google Scholar]
  • 68.Jellinger KA, Attems J. Does striatal pathology distinguish Parkinson disease with dementia and dementia with Lewy bodies? Acta Neuropathol. 2006;112:253–60. [DOI] [PubMed] [Google Scholar]
  • 69.Halliday GM, Song YJ, Harding AJ. Striatal β-amyloid in dementia with Lewy bodies but not Parkinson’s disease. J Neural Transm (Vienna). 2011;118:713–19. [DOI] [PubMed] [Google Scholar]
  • 70.Ruffmann C, Calboli FC, Bravi I, Gveric D, Curry LK, de Smith A, Pavlou S, Buxton JL, Blakemore AI, Takousis P, et al. Cortical Lewy bodies and Aβ burden are associated with prevalence and timing of dementia in Lewy body diseases. Neuropathol Appl Neurobiol. 2016;42:436–50. [DOI] [PubMed] [Google Scholar]
  • 71.Hepp DH, Vergoossen DL, Huisman E, Lemstra AW, Berendse HW, Rozemuller AJ, Foncke EM, van de Berg WD. Distribution and load of amyloid-β pathology in Parkinson disease and dementia with Lewy Bodies. J Neuropathol Exp Neurol. 2016;75:936–45. [DOI] [PubMed] [Google Scholar]
  • 72.Jellinger KA. Significance of cerebral amyloid angiopathy and other co-morbidities in Lewy body diseases. J Neural Transm (Vienna). 2021;128:687–99. [DOI] [PubMed] [Google Scholar]
  • 73.Jellinger KA. Morphological characteristics differentiate dementia with Lewy bodies from Parkinson disease with and without dementia. J Neural Transm (Vienna). 2023;130:891–904. [DOI] [PubMed] [Google Scholar]
  • 74.Jellinger KA, Korczyn AD. Are dementia with Lewy bodies and Parkinson’s disease dementia the same disease? BMC Med. 2018;16:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Boeve BF, Dickson DW, Duda JE, Ferman TJ, Galasko DR, Galvin JE, Goldman JG, Growdon JH, Hurtig HI, Kaufer DI, et al. Arguing against the proposed definition changes of PD. Mov Disord. 2016;31:1619–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Zaccai J, McCracken C, Brayne C. A systematic review of prevalence and incidence studies of dementia with Lewy bodies. Age Ageing. 2005;34:561–66. [DOI] [PubMed] [Google Scholar]
  • 77.Vann Jones SA, O’Brien. The prevalence and incidence of dementia with Lewy bodies: a systematic review of population and clinical studies. Psychol Med. 2014;44:673–83. [DOI] [PubMed] [Google Scholar]
  • 78.Hogan DB, Fiest KM, Roberts JI, Maxwell CJ, Dykeman J, Pringsheim T, Steeves T, Smith EE, Pearson D, Jetté N. The prevalence and incidence of dementia with Lewy Bodies: a systematic review. Can J Neurol Sci. 2016;43(Suppl, 1):S83–95. [DOI] [PubMed] [Google Scholar]
  • 79.Rizzo G, Arcuti S, Copetti M, Alessandria M, Savica R, Fontana A, Liguori R, Logroscino G. Accuracy of clinical diagnosis of dementia with Lewy bodies: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. 2018;89:358–66. [DOI] [PubMed] [Google Scholar]
  • 80.Tolosa E, Garrido A, Scholz SW, Poewe W. Challenges in the diagnosis of Parkinson’s disease. The Lancet Neurol. 2021;20:385–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Haroutunian V, Serby M, Purohit DP, Perl DP, Marin D, Lantz M, Mohs RC, Davis KL. Contribution of Lewy body inclusions to dementia in patients with and without Alzheimer disease neuropathological conditions. Arch Neurol. 2000;57:1145–50. [DOI] [PubMed] [Google Scholar]
  • 82.Schneider JA, Arvanitakis Z, Yu L, Boyle PA, Leurgans SE, Bennett DA. Cognitive impairment, decline and fluctuations in older community-dwelling subjects with Lewy bodies. Brain. 2012;135:3005–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Honda H, Sasaki K, Hamasaki H, Shijo M, Koyama S, Ohara T, Ninomiya T, Kiyohara Y, Suzuki SO, Iwaki T. Trends in autopsy-verified dementia prevalence over 29 years of the hisayama study. Neuropathology. 2016;36:383–87. [DOI] [PubMed] [Google Scholar]
  • 84.Dorsey ER, Sherer T, Okun MS, Bloem BR. The emerging evidence of the Parkinson pandemic. J Parkinsons Dis. 2018;8:S3–s8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sanghvi H, Singh R, Morrin H, Rajkumar AP. Systematic review of genetic association studies in people with Lewy body dementia. Int J Geriatr Psychiatry. 2020;35:436–48. [DOI] [PubMed] [Google Scholar]
  • 86.Zhao N, Liu CC, Qiao W, Bu G. Apolipoprotein E, receptors, and modulation of Alzheimer’s disease. Biol Psychiatry. 2018;83:347–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Moon HJ, Luo Y, Chugh D, Zhao L. Human apolipoprotein E glycosylation and sialylation: from structure to function. Front Mol Neurosci. 2024;17:1399965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Bras J, Guerreiro R, Darwent L, Parkkinen L, Ansorge O, Escott-Price V, Hernandez DG, Nalls MA, Clark LN, Honig LS, et al. Genetic analysis implicates APOE, SNCA and suggests lysosomal dysfunction in the etiology of dementia with Lewy bodies. Hum Mol Genet. 2014;23:6139–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Guerreiro R, Ross OA, Kun-Rodrigues C, Hernandez DG, Orme T, Eicher JD, Shepherd CE, Parkkinen L, Darwent L, Heckman MG, et al. Investigating the genetic architecture of dementia with Lewy bodies: a two-stage genome-wide association study. The Lancet Neurol. 2018;17:64–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Rongve A, Witoelar A, Ruiz A, Athanasiu L, Abdelnour C, Clarimon J, Heilmann-Heimbach S, Hernández I, Moreno-Grau S, de Rojas I, et al. GBA and APOE ε4 associate with sporadic dementia with Lewy bodies in European genome wide association study. Sci Rep. 2019;9:7013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Chia R, Sabir MS, Bandres-Ciga S, Saez-Atienzar S, Reynolds RH, Gustavsson E, Walton RL, Ahmed S, Viollet C, Ding J, et al. Genome sequencing analysis identifies new loci associated with Lewy body dementia and provides insights into its genetic architecture. Nat Genet. 2021;53:294–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Raulin AC, Doss SV, Trottier ZA, Ikezu TC, Bu G, Liu CC. ApoE in Alzheimer’s disease: pathophysiology and therapeutic strategies. Mol Neurodegener. 2022;17:72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Jackson RJ, Hyman BT, Serrano-Pozo A. Multifaceted roles of APOE in Alzheimer disease. Nat Rev Neurol. 2024;20:457–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Qu L, Xu S, Lan Z, Fang S, Xu Y, Zhu X. Apolipoprotein E in Alzheimer’s disease: focus on synaptic function and therapeutic strategy. Mol Neurobiol. 2024. [DOI] [PubMed]
  • 95.Raulin AC, Doss SV, Heckman MG, Craver EC, Li Z, Ikezu TC, Sekiya H, Liu CC, Martens YA, Rosenberg CL, et al. Impact of APOE on amyloid and tau accumulation in argyrophilic grain disease and Alzheimer’s disease. Acta Neuropathol Commun. 2024;12:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Dickson DW, Heckman MG, Murray ME, Soto AI, Walton RL, Diehl NN, van Gerpen JA, Uitti RJ, Wszolek ZK, Ertekin-Taner N, et al. APOE ε4 is associated with severity of Lewy body pathology independent of Alzheimer pathology. Neurology. 2018 1195;91:e1182–e. [DOI] [PMC free article] [PubMed]
  • 97.Tsuang D, Leverenz JB, Lopez OL, Hamilton RL, Bennett DA, Schneider JA, Buchman AS, Larson EB, Crane PK, Kaye JA, et al. APOE ε4 increases risk for dementia in pure synucleinopathies. JAMA Neurol. 2013;70:223–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kaivola K, Shah Z, Chia R, Scholz SW. Genetic evaluation of dementia with Lewy bodies implicates distinct disease subgroups. Brain. 2022;145:1757–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Real R, Martinez-Carrasco A, Reynolds RH, Lawton MA, Tan MMX, Shoai M, Corvol JC, Ryten M, Bresner C, Hubbard L, et al. Association between the LRP1B and APOE loci and the development of Parkinson’s disease dementia. Brain. 2023;146:1873–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Morley JF, Xie SX, Hurtig HI, Stern MB, Colcher A, Horn S, Dahodwala N, Duda JE, Weintraub D, Chen-Plotkin AS, et al. Genetic influences on cognitive decline in Parkinson’s disease. Mov Disord. 2012;27:512–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Jo S, Kim SO, Park KW, Lee SH, Hwang YS, Chung SJ. The role of APOE in cognitive trajectories and motor decline in Parkinson’s disease. Sci Rep. 2021;11:7819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Jung JH, Jeon S, Baik K, Lee YH, Chung SJ, Yoo HS, Jeong SH, Sohn YH, Lee PH, Ye BS. Apolipoprotein E4, amyloid, and cognition in Alzheimer’s and Lewy body disease. Neurobiol Aging. 2021;106:45–54. [DOI] [PubMed] [Google Scholar]
  • 103.Liu JY, Ma LZ, Wang J, Cui XJ, Sheng ZH, Fu Y, Li M, Ou YN, Yu JT, Tan L, Lian Y. Age-related association between APOE ɛ4 and cognitive progression in de novo Parkinson’s disease. J Alzheimers Dis. 2023;91:1121–32. [DOI] [PubMed] [Google Scholar]
  • 104.Zhao N, Attrebi ON, Ren Y, Qiao W, Sonustun B, Martens YA, Meneses AD, Li F, Shue F, Zheng J, et al. APOE4 exacerbates α-synuclein pathology and related toxicity independent of amyloid. Sci Transl Med. 2020;12. [DOI] [PMC free article] [PubMed]
  • 105.Davis AA, Inman CE, Wargel ZM, Dube U, Freeberg BM, Galluppi A, Haines JN, Dhavale DD, Miller R, Choudhury FA, et al. APOE genotype regulates pathology and disease progression in synucleinopathy. Sci Transl Med. 2020;12. [DOI] [PMC free article] [PubMed]
  • 106.Behl T, Kaur G, Fratila O, Buhas C, Judea-Pusta CT, Negrut N, Bustea C, Bungau S. Cross-talks among GBA mutations, glucocerebrosidase, and α-synuclein in GBA-associated Parkinson’s disease and their targeted therapeutic approaches: a comprehensive review. Transl NeurodeGener. 2021;10:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Clark LN, Kartsaklis LA, Wolf Gilbert R, Dorado B, Ross BM, Kisselev S, Verbitsky M, Mejia-Santana H, Cote LJ, Andrews H, et al. Association of glucocerebrosidase mutations with dementia with Lewy bodies. Arch Neurol. 2009;66:578–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Tsuang D, Leverenz JB, Lopez OL, Hamilton RL, Bennett DA, Schneider JA, Buchman AS, Larson EB, Crane PK, Kaye JA, et al. GBA mutations increase risk for Lewy body disease with and without Alzheimer disease pathology. Neurology. 2012;79:1944–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Creese B, Bell E, Johar I, Francis P, Ballard C, Aarsland D. Glucocerebrosidase mutations and neuropsychiatric phenotypes in Parkinson’s disease and Lewy body dementias: review and meta-analyses. Am J Med Genet B Neuropsychiatr Genet. 2018;177:232–41. [DOI] [PubMed] [Google Scholar]
  • 110.Blauwendraat C, Reed X, Krohn L, Heilbron K, Bandres-Ciga S, Tan M, Gibbs JR, Hernandez DG, Kumaran R, Langston R, et al. Genetic modifiers of risk and age at onset in GBA associated Parkinson’s disease and Lewy body dementia. Brain. 2020;143:234–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Gaubert S, Hourregue C, Mouton-Liger F, Millot P, Franco M, Amar-Bouaziz E, Aarsland D, Hugon J, Paquet C. Exploring the link between GBA1 mutations and dementia with Lewy bodies, a mini-review. Neurosci Biobehav Rev. 2022;141:104856. [DOI] [PubMed] [Google Scholar]
  • 112.Gündner AL, Duran-Pacheco G, Zimmermann S, Ruf I, Moors T, Baumann K, Jagasia R, van de Berg WDJ, Kremer T. Path mediation analysis reveals GBA impacts Lewy body disease status by increasing α-synuclein levels. Neurobiol Dis. 2019;121:205–13. [DOI] [PubMed] [Google Scholar]
  • 113.Nalls MA, Duran R, Lopez G, Kurzawa-Akanbi M, McKeith IG, Chinnery PF, Morris CM, Theuns J, Crosiers D, Cras P, et al. A multicenter study of glucocerebrosidase mutations in dementia with Lewy bodies. JAMA Neurol. 2013;70:727–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Liu G, Boot B, Locascio JJ, Jansen IE, Winder-Rhodes S, Eberly S, Elbaz A, Brice A, Ravina B, van Hilten JJ, et al. Specifically neuropathic Gaucher’s mutations accelerate cognitive decline in Parkinson’s. Ann Neurol. 2016;80:674–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Mata IF, Leverenz JB, Weintraub D, Trojanowski JQ, Chen-Plotkin A, Van Deerlin VM, Ritz B, Rausch R, Factor SA, Wood-Siverio C, et al. GBA variants are associated with a distinct pattern of cognitive deficits in Parkinson’s disease. Mov Disord. 2016;31:95–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Irwin DJ, Grossman M, Weintraub D, Hurtig HI, Duda JE, Xie SX, Lee EB, Van Deerlin VM, Lopez OL, Kofler JK, et al. Neuropathological and genetic correlates of survival and dementia onset in synucleinopathies: a retrospective analysis. The Lancet Neurol. 2017;16:55–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Walton RL, Koga S, Beasley AI, White LJ, Griesacker T, Murray ME, Kasanuki K, Hou X, Fiesel FC, Springer W, et al. Role of GBA variants in Lewy body disease neuropathology. Acta Neuropathol. 2024;147:54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Golbe LI, Di Iorio G, Bonavita V, Miller DC, Duvoisin RC. A large kindred with autosomal dominant Parkinson’s disease. Ann Neurol. 1990;27:276–82. [DOI] [PubMed] [Google Scholar]
  • 119.Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, Pike B, Root H, Rubenstein J, Boyer R, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science. 1997;276:2045–47. [DOI] [PubMed] [Google Scholar]
  • 120.Krüger R, Kuhn W, Müller T, Woitalla D, Graeber M, Kösel S, Przuntek H, Epplen JT, Schöls L, Riess O. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat Genet. 1998;18:106–08. [DOI] [PubMed] [Google Scholar]
  • 121.Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, Hulihan M, Peuralinna T, Dutra A, Nussbaum R, et al. Alpha-synuclein locus triplication causes Parkinson’s disease. Science. 2003;302:841. [DOI] [PubMed] [Google Scholar]
  • 122.Chartier-Harlin MC, Kachergus J, Roumier C, Mouroux V, Douay X, Lincoln S, Levecque C, Larvor L, Andrieux J, Hulihan M, et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet. 2004;364:1167–69. [DOI] [PubMed] [Google Scholar]
  • 123.Ibáñez P, Bonnet AM, Débarges B, Lohmann E, Tison F, Pollak P, Agid Y, Dürr A, Brice A. Causal relation between alpha-synuclein gene duplication and familial Parkinson’s disease. Lancet. 2004;364:1169–71. [DOI] [PubMed] [Google Scholar]
  • 124.Zarranz JJ, Alegre J, Gómez-Esteban JC, Lezcano E, Ros R, Ampuero I, Vidal L, Hoenicka J, Rodriguez O, Atarés B, et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol. 2004;55:164–73. [DOI] [PubMed] [Google Scholar]
  • 125.Ikeuchi T, Kakita A, Shiga A, Kasuga K, Kaneko H, Tan CF, Idezuka J, Wakabayashi K, Onodera O, Iwatsubo T, et al. Patients homozygous and heterozygous for SNCA duplication in a family with parkinsonism and dementia. Arch Neurol. 2008;65:514–19. [DOI] [PubMed] [Google Scholar]
  • 126.Seidel K, Schöls L, Nuber S, Petrasch-Parwez E, Gierga K, Wszolek Z, Dickson D, Gai WP, Bornemann A, Riess O, et al. First appraisal of brain pathology owing to A30P mutant alpha-synuclein. Ann Neurol. 2010;67:684–89. [DOI] [PubMed] [Google Scholar]
  • 127.Kiely AP, Asi YT, Kara E, Limousin P, Ling H, Lewis P, Proukakis C, Quinn N, Lees AJ, Hardy J, et al. α-Synucleinopathy associated with G51D SNCA Mutat: a link between Parkinson’s Disease and Multiple System Atrophy? Acta Neuropathol. 2013;125:753–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Appel-Cresswell S, Vilarino-Guell C, Encarnacion M, Sherman H, Yu I, Shah B, Weir D, Thompson C, Szu-Tu C, Trinh J, et al. Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson’s disease. Mov Disord. 2013;28:811–13. [DOI] [PubMed] [Google Scholar]
  • 129.Proukakis C, Dudzik CG, Brier T, MacKay DS, Cooper JM, Millhauser GL, Houlden H, Schapira AH. A novel α-synuclein missense mutation in Parkinson disease. Neurology. 2013;80:1062–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Kiely AP, Ling H, Asi YT, Kara E, Proukakis C, Schapira AH, Morris HR, Roberts HC, Lubbe S, Limousin P, et al. Distinct clinical and neuropathological features of G51D SNCA mutation cases compared with SNCA duplication and H50Q mutation. Mol Neurodegener. 2015;10:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Pasanen P, Myllykangas L, Siitonen M, Raunio A, Kaakkola S, Lyytinen J, Tienari PJ, Pöyhönen M, Paetau A. Novel α-synuclein mutation A53E associated with atypical multiple system atrophy and Parkinson’s disease-type pathology. Neurobiol Aging. 2014;35:2180. e2181–85. [DOI] [PubMed] [Google Scholar]
  • 132.Yoshino H, Hirano M, Stoessl AJ, Imamichi Y, Ikeda A, Li Y, Funayama M, Yamada I, Nakamura Y, Sossi V, et al. Homozygous alpha-synuclein p.A53V in familial Parkinson’s disease. Neurobiol Aging. 2017;57:.e248.247–248.212. [DOI] [PubMed] [Google Scholar]
  • 133.Kapasi A, Brosch JR, Nudelman KN, Agrawal S, Foroud TM, Schneider JA. A novel SNCA E83Q mutation in a case of dementia with Lewy bodies and atypical frontotemporal lobar degeneration. Neuropathology. 2020;40:620–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Fevga C, Park Y, Lohmann E, Kievit AJ, Breedveld GJ, Ferraro F, de Boer L, van Minkelen R, Hanagasi H, Boon A, et al. A new alpha-synuclein missense variant (Thr72Met) in two Turkish families with Parkinson’s disease. Parkinsonism Relat Disord. 2021;89:63–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Liu H, Koros C, Strohäker T, Schulte C, Bozi M, Varvaresos S, Ibáñez de Opakua A, Simitsi AM, Bougea A, Voumvourakis K, et al. A novel SNCA A30G mutation causes familial Parkinson’s disease. Mov Disord. 2021;36:1624–33. [DOI] [PubMed] [Google Scholar]
  • 136.Daida K, Shimonaka S, Shiba-Fukushima K, Ogata J, Yoshino H, Okuzumi A, Hatano T, Motoi Y, Hirunagi T, Katsuno M, et al. α-synuclein V15A variant in familial Parkinson’s disease exhibits a weaker lipid-binding property. Mov Disord. 2022;37:2075–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Duda JE, Giasson BI, Mabon ME, Miller DC, Golbe LI, Lee VM, Trojanowski JQ. Concurrence of alpha-synuclein and tau brain pathology in the contursi kindred. Acta Neuropathol. 2002;104:7–11. [DOI] [PubMed] [Google Scholar]
  • 138.Spira PJ, Sharpe DM, Halliday G, Cavanagh J, Nicholson GA. Clinical and pathological features of a parkinsonian syndrome in a family with an Ala53Thr alpha-synuclein mutation. Ann Neurol. 2001;49:313–19. [PubMed] [Google Scholar]
  • 139.Yamaguchi K, Cochran EJ, Murrell JR, Polymeropoulos MH, Shannon KM, Crowther RA, Goedert M, Ghetti B. Abundant neuritic inclusions and microvacuolar changes in a case of diffuse Lewy body disease with the A53T mutation in the alpha-synuclein gene. Acta Neuropathol. 2005;110:298–305. [DOI] [PubMed] [Google Scholar]
  • 140.Markopoulou K, Dickson DW, McComb RD, Wszolek ZK, Katechalidou L, Avery L, Stansbury MS, Chase BA. Clinical, neuropathological and genotypic variability in SNCA A53T familial Parkinson’s disease. Variability in familial Parkinson’s disease. Acta Neuropathol. 2008;116:25–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Nishioka K, Hashizume Y, Takanashi M, Daida K, Li Y, Yoshino H, Tambasco N, Prontera P, Hattori Y, Ueda A, et al. Pathological findings in a patient with alpha-synuclein p.A53T and familial Parkinson’s disease. Parkinsonism Relat Disord. 2020;81:183–87. [DOI] [PubMed] [Google Scholar]
  • 142.Lázaro DF, Rodrigues EF, Langohr R, Shahpasandzadeh H, Ribeiro T, Guerreiro P, Gerhardt E, Kröhnert K, Klucken J, Pereira MD, et al. Systematic comparison of the effects of alpha-synuclein mutations on its oligomerization and aggregation. PLoS Genet. 2014;10:e1004741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Tosatto L, Horrocks MH, Dear AJ, Knowles TP, Dalla Serra M, Cremades N, Dobson CM, Klenerman D. Single-molecule FRET studies on alpha-synuclein oligomerization of Parkinson’s disease genetically related mutants. Sci Rep. 2015;5:16696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Sierecki E, Giles N, Bowden Q, Polinkovsky ME, Steinbeck J, Arrioti N, Rahman D, Bhumkar A, Nicovich PR, Ross I, et al. Nanomolar oligomerization and selective co-aggregation of α-synuclein pathogenic mutants revealed by single-molecule fluorescence. Sci Rep. 2016;6:37630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Lima VA, Nascimento LA, Eliezer D, Follmer C. Role of Parkinson’s disease-linked mutations and N-Terminal acetylation on the oligomerization of α-Synuclein induced by 3, 4-dihydroxyphenylacetaldehyde. ACS Chem Neurosci. 2019;10:690–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Guella I, Evans DM, Szu-Tu C, Nosova E, Bortnick SF, Goldman JG, Dalrymple-Alford JC, Geurtsen GJ, Litvan I, Ross OA, et al. α-synuclein genetic variability: a biomarker for dementia in Parkinson disease. Ann Neurol. 2016;79:991–99. [DOI] [PubMed] [Google Scholar]
  • 147.Book A, Guella I, Candido T, Brice A, Hattori N, Jeon B, Farrer MJ. A meta-analysis of α-synuclein multiplication in familial parkinsonism. Front Neurol. 2018;9:1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Simitsi AM, Sfikas E, Koros C, Papagiannakis N, Beratis I, Papadimitriou D, Antonellou R, Fragiadaki S, Kontaxopoulou D, Picillo M, et al. Motor and nonmotor features of p.A53T alpha-synuclein PD vs idiopathic PD: longitudinal data from the PPMI study. J Neurol. 2025;272:203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hashimoto M, Rockenstein E, Mante M, Mallory M, Masliah E. Beta-Synuclein inhibits alpha-synuclein aggregation: a possible role as an anti-parkinsonian factor. Neuron. 2001;32:213–23. [DOI] [PubMed] [Google Scholar]
  • 150.Tsigelny IF, Bar-On P, Sharikov Y, Crews L, Hashimoto M, Miller MA, Keller SH, Platoshyn O, Yuan JX, Masliah E. Dynamics of alpha-synuclein aggregation and inhibition of pore-like oligomer development by beta-synuclein. The FEBS J. 2007;274:1862–77. [DOI] [PubMed] [Google Scholar]
  • 151.Beyer K, Domingo-Sàbat M, Santos C, Tolosa E, Ferrer I, Ariza A. The decrease of β-synuclein in cortical brain areas defines a molecular subgroup of dementia with Lewy bodies. Brain. 2010;133:3724–33. [DOI] [PubMed] [Google Scholar]
  • 152.Gámez-Valero A, Canet-Pons J, Urbizu A, Anillo A, Santos C, Ariza A, Beyer K. INDEL length and haplotypes in the β-Synuclein gene: a key to differentiate dementia with Lewy Bodies? J Alzheimers Dis. 2018;65:207–19. [DOI] [PubMed] [Google Scholar]
  • 153.Taschenberger G, Toloe J, Tereshchenko J, Akerboom J, Wales P, Benz R, Becker S, Outeiro TF, Looger LL, Bähr M, et al. β-synuclein aggregates and induces neurodegeneration in dopaminergic neurons. Ann Neurol. 2013;74:109–18. [DOI] [PubMed] [Google Scholar]
  • 154.Ohtake H, Limprasert P, Fan Y, Onodera O, Kakita A, Takahashi H, Bonner LT, Tsuang DW, Murray L IV VM, et al. Beta-synuclein gene alterations in dementia with Lewy bodies. Neurology. 2004;63:805–11. [DOI] [PMC free article] [PubMed]
  • 155.Psol M, Darvas SG, Leite K, Mahajani SU, Bähr M, Kügler S. Dementia with Lewy bodies-associated ß-synuclein mutations V70M and P123H cause mutation-specific neuropathological lesions. Hum Mol Genet. 2021;30:247–64. [DOI] [PubMed] [Google Scholar]
  • 156.Colom-Cadena M, Gelpi E, Martí MJ, Charif S, Dols-Icardo O, Blesa R, Clarimón J, Lleó A. MAPT H1 haplotype is associated with enhanced α-synuclein deposition in dementia with Lewy bodies. Neurobiol Aging. 2013;34:936–42. [DOI] [PubMed] [Google Scholar]
  • 157.Labbé C, Heckman MG, Lorenzo-Betancor O, Soto-Ortolaza AI, Walton RL, Murray ME, Allen M, Uitti RJ, Wszolek ZK, Smith GE, et al. MAPT haplotype H1G is associated with increased risk of dementia with Lewy bodies. Alzheimers Dement. 2016;12:1297–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Heckman MG, Kasanuki K, Brennan RR, Labbé C, Vargas ER, Soto AI, Murray ME, Koga S, Dickson DW, Ross OA. Association of MAPT H1 subhaplotypes with neuropathology of Lewy body disease. Mov Disord. 2019;34:1325–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Dickson DW. Dementia with Lewy bodies: neuropathology. J Geriatr Psychiatry Neurol. 2002;15:210–16. [DOI] [PubMed] [Google Scholar]
  • 160.Dickson DW, Braak H, Duda JE, Duyckaerts C, Gasser T, Halliday GM, Hardy J, Leverenz JB, Del Tredici K, Wszolek ZK, Litvan I. Neuropathological assessment of Parkinson’s disease: refining the diagnostic criteria. The Lancet Neurol. 2009;8:1150–57. [DOI] [PubMed] [Google Scholar]
  • 161.Dickson DW. Parkinson’s disease and parkinsonism: neuropathology. Cold Spring Harb Perspect Med. 2012;2. [DOI] [PMC free article] [PubMed]
  • 162.Dickson DW. Neuropathology of Parkinson disease. Parkinsonism Relat Disord. 2018;46(Suppl, 1):S30–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Koga S, Sekiya H, Kondru N, Ross OA, Dickson DW. Neuropathology and molecular diagnosis of synucleinopathies. Mol Neurodegener. 2021;16:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Duffy PE, Tennyson VM. Phase and electron microscopic observations of Lewy bodies and melanin granules in the substantia nigra and locus caeruleus in Parkinson’s disease. J Neuropathol & Exp Neurol. 1965;24:398–414. [Google Scholar]
  • 165.Fujiwara H, Hasegawa M, Dohmae N, Kawashima A, Masliah E, Goldberg MS, Shen J, Takio K, Iwatsubo T. Alpha-synuclein is phosphorylated in synucleinopathy lesions. Nat Cell Biol. 2002;4:160–64. [DOI] [PubMed] [Google Scholar]
  • 166.Kuzuhara S, Mori H, Izumiyama N, Yoshimura M, Ihara Y. Lewy bodies are ubiquitinated. A light and electron microscopic immunocytochemical study. Acta Neuropathol. 1988;75:345–53. [DOI] [PubMed] [Google Scholar]
  • 167.Kuusisto E, Salminen A, Alafuzoff I. Ubiquitin-binding protein p62 is present in neuronal and glial inclusions in human tauopathies and synucleinopathies. Neuroreport. 2001;12:2085–90. [DOI] [PubMed] [Google Scholar]
  • 168.Kuusisto E, Parkkinen L, Alafuzoff I. Morphogenesis of Lewy bodies: dissimilar incorporation of alpha-synuclein, ubiquitin, and p62. J Neuropathol Exp Neurol. 2003;62:1241–53. [DOI] [PubMed] [Google Scholar]
  • 169.Pollanen MS, Bergeron C, Weyer L. Detergent-insoluble cortical Lewy body fibrils share epitopes with neurofilament and tau. J Neurochem. 1992;58:1953–56. [DOI] [PubMed] [Google Scholar]
  • 170.Pollanen MS, Bergeron C, Weyer L. Characterization of a shared epitope in cortical Lewy body fibrils and Alzheimer paired helical filaments. Acta Neuropathol. 1994;88:1–6. [DOI] [PubMed] [Google Scholar]
  • 171.Leverenz JB, Umar I, Wang Q, Montine TJ, McMillan PJ, Tsuang DW, Jin J, Pan C, Shin J, Zhu D, Zhang J. Proteomic identification of novel proteins in cortical Lewy bodies. Brain Pathol. 2007;17:139–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Xia Q, Liao L, Cheng D, Duong DM, Gearing M, Lah JJ, Levey AI, Peng J. Proteomic identification of novel proteins associated with Lewy bodies. Front Biosci. 2008;13:3850–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Fearnley JM, Lees AJ. Ageing and Parkinson’s disease: substantia nigra regional selectivity. Brain. 1991;114(Pt 5):2283–301. [DOI] [PubMed] [Google Scholar]
  • 174.Lewy F, Lewandowsky M. Handbuch der Neurologie. Berlin: Julius Springer. 1912.
  • 175.Wakabayashi K, Takahashi H, Obata K, Ikuta F. Immunocytochemical localization of synaptic vesicle-specific protein in Lewy body-containing neurons in Parkinson’s disease. Neurosci Lett. 1992;138:237–40. [DOI] [PubMed] [Google Scholar]
  • 176.Dale GE, Probst A, Luthert P, Martin J, Anderton BH, Leigh PN. Relationships between Lewy bodies and pale bodies in Parkinson’s disease. Acta Neuropathol. 1992;83:525–29. [DOI] [PubMed] [Google Scholar]
  • 177.Okazaki H, Lipkin LE, Aronson SM. Diffuse intracytoplasmic ganglionic inclusions (Lewy type) associated with progressive dementia and quadriparesis in flexion. J Neuropathol Exp Neurol. 1961;20:237–44. [DOI] [PubMed] [Google Scholar]
  • 178.Dickson DW, Ruan D, Crystal H, Mark MH, Davies P, Kress Y, Yen SH. Hippocampal degeneration differentiates diffuse Lewy body disease (DLBD) from Alzheimer’s disease: light and electron microscopic immunocytochemistry of CA2-3 neurites specific to DLBD. Neurology. 1991;41:1402–09. [DOI] [PubMed] [Google Scholar]
  • 179.McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman H, Cummings J, Duda JE, Lippa C, Perry EK, et al. Diagnosis and management of dementia with Lewy bodies: third report of the DLB consortium. Neurology. 2005;65:1863–72. [DOI] [PubMed] [Google Scholar]
  • 180.Kanazawa T, Uchihara T, Takahashi A, Nakamura A, Orimo S, Mizusawa H. Three-layered structure shared between Lewy bodies and Lewy neurites-three-dimensional reconstruction of triple-labeled sections. Brain Pathol. 2008;18:415–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Braak H, Del Tredici K, Rüb U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211. [DOI] [PubMed] [Google Scholar]
  • 182.Beach TG, Adler CH, Sue LI, Vedders L, Lue L, White Iii CL, Akiyama H, Caviness JN, Shill HA, Sabbagh MN, Walker DG. Multi-organ distribution of phosphorylated alpha-synuclein histopathology in subjects with Lewy body disorders. Acta Neuropathol. 2010;119:689–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Tanei ZI, Saito Y, Ito S, Matsubara T, Motoda A, Yamazaki M, Sakashita Y, Kawakami I, Ikemura M, Tanaka S, et al. Lewy pathology of the esophagus correlates with the progression of Lewy body disease: a Japanese cohort study of autopsy cases. Acta Neuropathol. 2021;141:25–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Matsubara T, Kameyama M, Tanaka N, Sengoku R, Orita M, Furuta K, Iwata A, Arai T, Maruyama H, Saito Y, Murayama S. Autopsy validation of the diagnostic accuracy of (123)I-Metaiodobenzylguanidine myocardial scintigraphy for Lewy Body Disease. Neurology. 2022;98:e1648–e 1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Hansen LA, Masliah E, Terry RD, Mirra SS. A neuropathological subset of Alzheimer’s disease with concomitant Lewy body disease and spongiform change. Acta Neuropathol. 1989;78:194–201. [DOI] [PubMed] [Google Scholar]
  • 186.Fujino Y, Dickson DW. Limbic lobe microvacuolation is minimal in Alzheimer’s disease in the absence of concurrent Lewy body disease. Int J Clin Exp Pathol. 2008;1:369–75. [PMC free article] [PubMed] [Google Scholar]
  • 187.Iseki E, Li F, Odawara T, Kosaka K. Hippocampal pathology in diffuse Lewy body disease using ubiquitin immunohistochemistry. J Neurol Sci. 1997;149:165–69. [DOI] [PubMed] [Google Scholar]
  • 188.Sherzai A, Edland SD, Masliah E, Hansen L, Pizzo DP, Sherzai A, Corey-Bloom J. Spongiform change in dementia with Lewy bodies and Alzheimer disease. Alzheimer Dis Assoc Disord. 2013;27:157–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Budka H, Aguzzi A, Brown P, Brucher JM, Bugiani O, Gullotta F, Haltia M, Hauw JJ, Ironside JW, Jellinger K, et al. Neuropathological diagnostic criteria for Creutzfeldt-Jakob disease (CJD) and other human spongiform encephalopathies (prion diseases). Brain Pathol. 1995;5:459–66. [DOI] [PubMed] [Google Scholar]
  • 190.Kosaka K, Yoshimura M, Ikeda K, Budka H. Diffuse type of Lewy body disease: progressive dementia with abundant cortical Lewy bodies and senile changes of varying degree-a new disease? Clin neuropathol. 1984;3:185–92. [PubMed] [Google Scholar]
  • 191.Beach TG, Adler CH, Lue L, Sue LI, Bachalakuri J, Henry-Watson J, Sasse J, Boyer S, Shirohi S, Brooks R, et al. Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta Neuropathol. 2009;117:613–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Saito Y, Kawashima A, Ruberu NN, Fujiwara H, Koyama S, Sawabe M, Arai T, Nagura H, Yamanouchi H, Hasegawa M, et al. Accumulation of phosphorylated alpha-synuclein in aging human brain. J Neuropathol Exp Neurol. 2003;62:644–54. [DOI] [PubMed] [Google Scholar]
  • 193.Uchikado H, Lin WL, DeLucia MW, Dickson DW. Alzheimer disease with amygdala Lewy bodies: a distinct form of alpha-synucleinopathy. J Neuropathol Exp Neurol. 2006;65:685–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Iseki E, Marui W, Kosaka K, Uéda K. Frequent coexistence of Lewy bodies and neurofibrillary tangles in the same neurons of patients with diffuse Lewy body disease. Neurosci Lett. 1999;265:9–12. [DOI] [PubMed] [Google Scholar]
  • 195.Ishizawa T, Mattila P, Davies P, Wang D, Dickson DW. Colocalization of tau and alpha-synuclein epitopes in Lewy bodies. J Neuropathol Exp Neurol. 2003;62:389–97. [DOI] [PubMed] [Google Scholar]
  • 196.Fujishiro H, Tsuboi Y, Lin WL, Uchikado H, Dickson DW. Co-localization of tau and alpha-synuclein in the olfactory bulb in Alzheimer’s disease with amygdala Lewy bodies. Acta Neuropathol. 2008;116:17–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Leverenz JB, Hamilton R, Tsuang DW, Schantz A, Vavrek D, Larson EB, Kukull WA, Lopez O, Galasko D, Masliah E, et al. Empiric refinement of the pathologic assessment of Lewy-related pathology in the dementia patient. Brain Pathol. 2008;18:220–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Alafuzoff I, Ince PG, Arzberger T, Al-Sarraj S, Bell J, Bodi I, Bogdanovic N, Bugiani O, Ferrer I, Gelpi E, et al. Staging/Typing of Lewy body related alpha-synuclein pathology: a study of the BrainNet Europe consortium. Acta Neuropathol. 2009;117:635–52. [DOI] [PubMed] [Google Scholar]
  • 199.Attems J, Toledo JB, Walker L, Gelpi E, Gentleman S, Halliday G, Hortobagyi T, Jellinger K, Kovacs GG, Lee EB, et al. Neuropathological consensus criteria for the evaluation of Lewy pathology in post-mortem brains: a multi-centre study. Acta Neuropathol. 2021;141:159–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Kordower JH, Olanow CW, Dodiya HB, Chu Y, Beach TG, Adler CH, Halliday GM, Bartus RT. Disease duration and the integrity of the nigrostriatal system in Parkinson’s disease. Brain. 2013;136:2419–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Dickson DW, Fujishiro H, DelleDonne A, Menke J, Ahmed Z, Klos KJ, Josephs KA, Frigerio R, Burnett M, Parisi JE, Ahlskog JE. Evidence that incidental Lewy body disease is pre-symptomatic Parkinson’s disease. Acta Neuropathol. 2008;115:437–44. [DOI] [PubMed] [Google Scholar]
  • 202.Koeglsperger T, Rumpf SL, Schließer P, Struebing FL, Brendel M, Levin J, Trenkwalder C, Höglinger GU, Herms J. Neuropathology of incidental Lewy body & prodromal Parkinson’s disease. Mol NeurodeGener. 2023;18:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Gelpi E, Navarro-Otano J, Tolosa E, Gaig C, Compta Y, Rey MJ, Martí MJ, Hernández I, Valldeoriola F, Reñé R, Ribalta T. Multiple organ involvement by alpha-synuclein pathology in Lewy body disorders. Mov Disord. 2014;29:1010–18. [DOI] [PubMed] [Google Scholar]
  • 204.Ito S, Takao M, Hatsuta H, Kanemaru K, Arai T, Saito Y, Fukayama M, Murayama S. Alpha-synuclein immunohistochemistry of gastrointestinal and biliary surgical specimens for diagnosis of Lewy body disease. Int J Clin Exp Pathol. 2014;7:1714–23. [PMC free article] [PubMed] [Google Scholar]
  • 205.Jotanovic J, Milin-Lazovic J, Alafuzoff I. Gastrointestinal biopsy obtained during cancer screening, a biological Marker for α-synucleinopathy? J Neuropathol Exp Neurol. 2022;81:356–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Beach TG, Adler CH, Serrano G, Sue LI, Walker DG, Dugger BN, Shill HA, Driver-Dunckley E, Caviness JN, Intorcia A, et al. Prevalence of submandibular gland synucleinopathy in Parkinson’s disease, dementia with Lewy bodies and other Lewy body disorders. J Parkinsons Dis. 2016;6:153–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Sakashita Y, Matsubara T, Takata T, Tanei ZI, Motoda A, Yamazaki M, Kawakami I, Sengoku R, Saito Y, Arai T, et al. Lewy pathology of the submandibular gland in Lewy body disease: a report of autopsy cases. Neuropathology. 2021;41:476–83. [DOI] [PubMed] [Google Scholar]
  • 208.Ikemura M, Saito Y, Sengoku R, Sakiyama Y, Hatsuta H, Kanemaru K, Sawabe M, Arai T, Ito G, Iwatsubo T, et al. Lewy body pathology involves cutaneous nerves. J Neuropathol Exp Neurol. 2008;67:945–53. [DOI] [PubMed] [Google Scholar]
  • 209.Donadio V, Incensi A, Rizzo G, Capellari S, Pantieri R, Stanzani Maserati M, Devigili G, Eleopra R, Defazio G, Montini F, et al. A new potential biomarker for dementia with Lewy bodies: skin nerve α-synuclein deposits. Neurology. 2017;89:318–26. [DOI] [PubMed] [Google Scholar]
  • 210.Gibbons CH, Levine T, Adler C, Bellaire B, Wang N, Stohl J, Agarwal P, Aldridge GM, Barboi A, Evidente VGH, et al. Skin biopsy detection of phosphorylated α-synuclein in patients with synucleinopathies. JAMA. 2024;331:1298–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Iwanaga K, Wakabayashi K, Yoshimoto M, Tomita I, Satoh H, Takashima H, Satoh A, Seto M, Tsujihata M, Takahashi H. Lewy body-type degeneration in cardiac plexus in Parkinson’s and incidental Lewy body diseases. Neurology. 1999;52:1269–71. [DOI] [PubMed] [Google Scholar]
  • 212.Orimo S, Uchihara T, Nakamura A, Mori F, Kakita A, Wakabayashi K, Takahashi H. Axonal alpha-synuclein aggregates herald centripetal degeneration of cardiac sympathetic nerve in Parkinson’s disease. Brain. 2008;131:642–50. [DOI] [PubMed] [Google Scholar]
  • 213.Fumimura Y, Ikemura M, Saito Y, Sengoku R, Kanemaru K, Sawabe M, Arai T, Ito G, Iwatsubo T, Fukayama M, et al. Analysis of the adrenal gland is useful for evaluating pathology of the peripheral autonomic nervous system in Lewy body disease. J Neuropathol Exp Neurol. 2007;66:354–62. [DOI] [PubMed] [Google Scholar]
  • 214.Raunio A, Kivistö V, Kero M, Tuimala J, Savola S, Oinas M, Kok E, Colangelo K, Paetau A, Polvikoski T, et al. Distribution of Lewy-related pathology in the brain, spinal cord, and periphery: the population-based Vantaa 85 + study. Acta Neuropathol Commun. 2022;10:178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Yuan X, Nie S, Yang Y, Liu C, Xia D, Meng L, Xia Y, Su H, Zhang C, Bu L, et al. Propagation of pathologic α-synuclein from kidney to brain may contribute to Parkinson’s disease. Nat Neurosci. 2025;28:577–88. [DOI] [PubMed] [Google Scholar]
  • 216.de Ruyter Fj H, Evers M, Morrema THJ, Dijkstra AA, den Haan J, Twisk JWR, de Boer Jf, Scheltens P, Bouwman FH, Verbraak FD, et al. Neuropathological hallmarks in the post-mortem retina of neurodegenerative diseases. Acta Neuropathol. 2024;148:24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Cabin DE, Shimazu K, Murphy D, Cole NB, Gottschalk W, McIlwain KL, Orrison B, Chen A, Ellis CE, Paylor R, et al. Synaptic vesicle depletion correlates with attenuated synaptic responses to prolonged repetitive stimulation in mice lacking alpha-synuclein. J Neurosci. 2002;22:8797–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Chadchankar H, Ihalainen J, Tanila H, Yavich L. Decreased reuptake of dopamine in the dorsal striatum in the absence of α-synuclein. Brain Res. 2011;1382:37–44. [DOI] [PubMed] [Google Scholar]
  • 219.Nemani VM, Lu W, Berge V, Nakamura K, Onoa B, Lee MK, Chaudhry FA, Nicoll RA, Edwards RH. Increased expression of alpha-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron. 2010;65:66–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Scott DA, Tabarean I, Tang Y, Cartier A, Masliah E, Roy S. A pathologic cascade leading to synaptic dysfunction in alpha-synuclein-induced neurodegeneration. J Neurosci. 2010;30:8083–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Garcia-Reitböck P, Anichtchik O, Bellucci A, Iovino M, Ballini C, Fineberg E, Ghetti B, Della Corte L, Spano P, Tofaris GK, et al. SNARE protein redistribution and synaptic failure in a transgenic mouse model of Parkinson’s disease. Brain. 2010;133:2032–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Brolin E, Ingelsson M, Bergström J, Erlandsson A. Altered distribution of SNARE proteins in primary neurons exposed to different alpha-synuclein proteoforms. Cell Mol Neurobiol. 2023;43:3023–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Román-Vendrell C, Medeiros AT, Sanderson JB, Jiang H, Bartels T, Morgan JR. Effects of excess brain-derived human α-synuclein on synaptic vesicle trafficking. Front Neurosci. 2021;15:639414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Fanning S, Selkoe D, Dettmer U. Vesicle trafficking and lipid metabolism in synucleinopathy. Acta Neuropathol. 2021;141:491–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Phan JA, Stokholm K, Zareba-Paslawska J, Jakobsen S, Vang K, Gjedde A, Landau AM, Romero-Ramos M. Early synaptic dysfunction induced by α-synuclein in a rat model of Parkinson’s disease. Sci Rep. 2017;7:6363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Longhena F, Faustini G, Missale C, Pizzi M, Bellucci A. Dopamine transporter/α-synuclein complexes are altered in the post mortem caudate putamen of Parkinson’s disease: an in situ proximity ligation assay study. Int J Mol Sci. 2018;19. [DOI] [PMC free article] [PubMed]
  • 227.Tozzi A, Sciaccaluga M, Loffredo V, Megaro A, Ledonne A, Cardinale A, Federici M, Bellingacci L, Paciotti S, Ferrari E, et al. Dopamine-dependent early synaptic and motor dysfunctions induced by α-synuclein in the nigrostriatal circuit. Brain. 2021;144:3477–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Chu Y, Morfini GA, Langhamer LB, He Y, Brady ST, Kordower JH. Alterations in axonal transport motor proteins in sporadic and experimental Parkinson’s disease. Brain. 2012;135:2058–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Koch JC, Bitow F, Haack J, d’Hedouville Z, Zhang JN, Tönges L, Michel U, Oliveira LM, Jovin TM, Liman J, et al. Alpha-Synuclein affects neurite morphology, autophagy, vesicle transport and axonal degeneration in CNS neurons. Cell Death Dis. 2015;6:e 1811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Mazzulli JR, Zunke F, Isacson O, Studer L, Krainc D. α-synuclein-induced lysosomal dysfunction occurs through disruptions in protein trafficking in human midbrain synucleinopathy models. Proc Natl Acad Sci USA. 2016;113:1931–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Tang Q, Gao P, Arzberger T, Höllerhage M, Herms J, Höglinger G, Koeglsperger T. Alpha-Synuclein defects autophagy by impairing SNAP29-mediated autophagosome-lysosome fusion. Cell Death Dis. 2021;12:854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Zhang NY, Tang Z, Liu CW. Alpha-Synuclein protofibrils inhibit 26 S proteasome-mediated protein degradation: understanding the cytotoxicity of protein protofibrils in neurodegenerative disease pathogenesis. J Biol Chem. 2008;283:20288–98. [DOI] [PubMed] [Google Scholar]
  • 233.McKinnon C, De Snoo ML, Gondard E, Neudorfer C, Chau H, Ngana SG, O’Hara DM, Brotchie JM, Koprich JB, Lozano AM, et al. Early-onset impairment of the ubiquitin-proteasome system in dopaminergic neurons caused by α-synuclein. Acta Neuropathol Commun. 2020;8:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Wang X, Becker K, Levine N, Zhang M, Lieberman AP, Moore DJ, Ma J. Pathogenic alpha-synuclein aggregates preferentially bind to mitochondria and affect cellular respiration. Acta Neuropathol Commun. 2019;7:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Lurette O, Martín-Jiménez R, Khan M, Sheta R, Jean S, Schofield M, Teixeira M, Rodriguez-Aller R, Perron I, Oueslati A, Hebert-Chatelain E. Aggregation of alpha-synuclein disrupts mitochondrial metabolism and induce mitophagy via cardiolipin externalization. Cell Death Dis. 2023;14:729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Geibl FF, Henrich MT, Xie Z, Zampese E, Ueda J, Tkatch T, Wokosin DL, Nasiri E, Grotmann CA, Dawson VL, et al. α-Synuclein pathology disrupts mitochondrial function in dopaminergic and cholinergic neurons at-risk in Parkinson’s disease. Mol NeurodeGener. 2024;19:69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Freeman D, Cedillos R, Choyke S, Lukic Z, McGuire K, Marvin S, Burrage AM, Sudholt S, Rana A, O’Connor C, et al. Alpha-synuclein induces lysosomal rupture and cathepsin dependent reactive oxygen species following endocytosis. PLoS One. 2013;8:e62143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Dai L, Liu M, Ke W, Chen L, Fang X, Zhang Z. Lysosomal dysfunction in α-synuclein pathology: Molecular mechanisms and therapeutic strategies. Cell Mol Life Sci. 2024;81:382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Mahul-Mellier AL, Burtscher J, Maharjan N, Weerens L, Croisier M, Kuttler F, Leleu M, Knott GW, Lashuel HA. The process of Lewy body formation, rather than simply α-synuclein fibrillization, is one of the major drivers of neurodegeneration. Proc Natl Acad Sci USA. 2020;117:4971–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Sarkar S, Olsen AL, Sygnecka K, Lohr KM. Feany MB. α-synuclein impairs autophagosome maturation through abnormal actin stabilization. PLoS Genet. 2021;17:e1009359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Choi YR, Park SJ, Park SM. Molecular events underlying the cell-to-cell transmission of α-synuclein. The FEBS J. 2021;288:6593–602. [DOI] [PubMed] [Google Scholar]
  • 242.Tofaris GK. Initiation and progression of α-synuclein pathology in Parkinson’s disease. Cell Mol Life Sci. 2022;79:210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Sharon R, Bar-Joseph I, Frosch MP, Walsh DM, Hamilton JA, Selkoe DJ. The formation of highly soluble oligomers of alpha-synuclein is regulated by fatty acids and enhanced in Parkinson’s disease. Neuron. 2003;37:583–95. [DOI] [PubMed] [Google Scholar]
  • 244.Cascella R, Bigi A, Cremades N, Cecchi C. Effects of oligomer toxicity, fibril toxicity and fibril spreading in synucleinopathies. Cell Mol Life Sci. 2022;79:174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Olanow CW, Perl DP, DeMartino GN, McNaught KS. Lewy-body formation is an aggresome-related process: a hypothesis. The Lancet Neurol. 2004;3:496–503. [DOI] [PubMed] [Google Scholar]
  • 246.Wakabayashi K, Tanji K, Odagiri S, Miki Y, Mori F, Takahashi H. The Lewy body in Parkinson’s disease and related neurodegenerative disorders. Mol Neurobiol. 2013;47:495–508. [DOI] [PubMed] [Google Scholar]
  • 247.Chartier S, Duyckaerts C. Is Lewy pathology in the human nervous system chiefly an indicator of neuronal protection or of toxicity? Cell Tissue Res. 2018;373:149–60. [DOI] [PubMed] [Google Scholar]
  • 248.Chen L, Feany MB. Alpha-synuclein phosphorylation controls neurotoxicity and inclusion formation in a drosophila model of Parkinson disease. Nat Neurosci. 2005;8:657–63. [DOI] [PubMed] [Google Scholar]
  • 249.Sultana Z, Paleologou KE, Al-Mansoori KM, Ardah MT, Singh N, Usmani S, Jiao H, Martin FL, Bharath MM, Vali S, El-Agnaf OM. Dynamic modeling of α-synuclein aggregation in dopaminergic neuronal system indicates points of neuroprotective intervention: experimental validation with implications for Parkinson’s therapy. Neuroscience. 2011;199:303–17. [DOI] [PubMed] [Google Scholar]
  • 250.Kuznetsov AV. Criterion for assessing accumulated neurotoxicity of alpha-synuclein oligomers in Parkinson’s disease. Int J Numer Method Biomed Eng. 2025;41:e70027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Ferman TJ, Aoki N, Crook JE, Murray ME, Graff-Radford NR, van Gerpen JA, Uitti RJ, Wszolek ZK, Graff-Radford J, Pedraza O, et al. The limbic and neocortical contribution of α-synuclein, tau, and amyloid β to disease duration in dementia with Lewy bodies. Alzheimers DeMent. 2018;14:330–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Graff-Radford J, Aakre J, Savica R, Boeve B, Kremers WK, Ferman TJ, Jones DT, Kantarci K, Knopman DS, Dickson DW, et al. Duration and pathologic correlates of Lewy body disease. JAMA Neurol. 2017;74:310–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Adler CH, Beach TG, Zhang N, Shill HA, Driver-Dunckley E, Caviness JN, Mehta SH, Sabbagh MN, Serrano GE, Sue LI, et al. Unified staging system for Lewy body disorders: clinicopathologic correlations and comparison to Braak staging. J Neuropathol Exp Neurol. 2019;78:891–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Hurtig HI, Trojanowski JQ, Galvin J, Ewbank D, Schmidt ML, Lee VM, Clark CM, Glosser G, Stern MB, Gollomp SM, Arnold SE. Alpha-synuclein cortical Lewy bodies correlate with dementia in Parkinson’s disease. Neurology. 2000;54:1916–21. [DOI] [PubMed] [Google Scholar]
  • 255.Apaydin H, Ahlskog JE, Parisi JE, Boeve BF, Dickson DW. Parkinson disease neuropathology: later-developing dementia and loss of the levodopa response. Arch Neurol. 2002;59:102–12. [DOI] [PubMed] [Google Scholar]
  • 256.Kempster PA, O’Sullivan SS, Holton JL, Revesz T, Lees AJ. Relationships between age and late progression of Parkinson’s disease: a clinico-pathological study. Brain. 2010;133:1755–62. [DOI] [PubMed] [Google Scholar]
  • 257.Irwin DJ, White MT, Toledo JB, Xie SX, Robinson JL, Van Deerlin V, Lee VM, Leverenz JB, Montine TJ, Duda JE, et al. Neuropathologic substrates of Parkinson disease dementia. Ann Neurol. 2012;72:587–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Horvath J, Herrmann FR, Burkhard PR, Bouras C, Kövari E. Neuropathology of dementia in a large cohort of patients with Parkinson’s disease. Parkinsonism Relat Disord. 2013;19:864–68; discussion 864. [DOI] [PubMed] [Google Scholar]
  • 259.Galvin JE, Pollack J, Morris JC. Clinical phenotype of Parkinson disease dementia. Neurology. 2006;67:1605–11. [DOI] [PubMed] [Google Scholar]
  • 260.Libow LS, Frisina PG, Haroutunian V, Perl DP, Purohit DP. Parkinson’s disease dementia: a diminished role for the Lewy body. Parkinsonism Relat Disord. 2009;15:572–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Colosimo C, Hughes AJ, Kilford L, Lees AJ. Lewy body cortical involvement may not always predict dementia in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2003;74:852–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Parkkinen L, Kauppinen T, Pirttilä T, Autere JM, Alafuzoff I. Alpha-synuclein pathology does not predict extrapyramidal symptoms or dementia. Ann Neurol. 2005;57:82–91. [DOI] [PubMed] [Google Scholar]
  • 263.Greffard S, Verny M, Bonnet AM, Seilhean D, Hauw JJ. Duyckaerts C. A stable proportion of Lewy body bearing neurons in the substantia nigra suggests a model in which the Lewy body causes neuronal death. Neurobiol Aging. 2010;31:99–103. [DOI] [PubMed] [Google Scholar]
  • 264.Mattila PM, Röyttä M, Torikka H, Dickson DW, Rinne JO. Cortical Lewy bodies and alzheimer-type changes in patients with Parkinson’s disease. Acta Neuropathol. 1998;95:576–82. [DOI] [PubMed] [Google Scholar]
  • 265.Mattila PM, Rinne JO, Helenius H, Dickson DW, Röyttä M. Alpha-synuclein-immunoreactive cortical Lewy bodies are associated with cognitive impairment in Parkinson’s disease. Acta Neuropathol. 2000;100:285–90. [DOI] [PubMed] [Google Scholar]
  • 266.Braak H, Rüb U, Jansen Steur EN, Del Tredici K, de Vos RA. Cognitive status correlates with neuropathologic stage in Parkinson disease. Neurology. 2005;64:1404–10. [DOI] [PubMed] [Google Scholar]
  • 267.Howlett DR, Whitfield D, Johnson M, Attems J, O’Brien JT, Aarsland D, Lai MK, Lee JH, Chen C, Ballard C, et al. Regional multiple pathology scores are associated with cognitive decline in Lewy body dementias. Brain Pathol. 2015;25:401–08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Aarsland D, Perry R, Brown A, Larsen JP, Ballard C. Neuropathology of dementia in Parkinson’s disease: a prospective, community-based study. Ann Neurol. 2005;58:773–76. [DOI] [PubMed] [Google Scholar]
  • 269.Merdes AR, Hansen LA, Jeste DV, Galasko D, Hofstetter CR, Ho GJ, Thal LJ, Corey-Bloom J. Influence of Alzheimer pathology on clinical diagnostic accuracy in dementia with Lewy bodies. Neurology. 2003;60:1586–90. [DOI] [PubMed] [Google Scholar]
  • 270.Ballard CG, Jacoby R, Del Ser T, Khan MN, Munoz DG, Holmes C, Nagy Z, Perry EK, Joachim C, Jaros E, et al. Neuropathological substrates of psychiatric symptoms in prospectively studied patients with autopsy-confirmed dementia with Lewy bodies. Am J Psychiatry. 2004;161:843–49. [DOI] [PubMed] [Google Scholar]
  • 271.Tsuang D, Larson EB, Bolen E, Thompson ML, Peskind E, Bowen J, McCormick W, Teri L, Kukull W, Vavrek D, et al. Visual hallucinations in dementia: a prospective community-based study with autopsy. Am J Geriatr Psychiatry. 2009;17:317–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Ferman TJ, Arvanitakis Z, Fujishiro H, Duara R, Parfitt F, Purdy M, Waters C, Barker W, Graff-Radford NR, Dickson DW. Pathology and temporal onset of visual hallucinations, misperceptions and family misidentification distinguishes dementia with Lewy bodies from Alzheimer’s disease. Parkinsonism Relat Disord. 2013;19:227–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Harding AJ, Broe GA, Halliday GM. Visual hallucinations in Lewy body disease relate to Lewy bodies in the temporal lobe. Brain. 2002;125:391–403. [DOI] [PubMed] [Google Scholar]
  • 274.Yamamoto R, Iseki E, Murayama N, Minegishi M, Marui W, Togo T, Katsuse O, Kato M, Iwatsubo T, Kosaka K, Arai H. Investigation of Lewy pathology in the visual pathway of brains of dementia with Lewy bodies. J Neurol Sci. 2006;246:95–101. [DOI] [PubMed] [Google Scholar]
  • 275.Ferman TJ, Smith GE, Boeve BF, Ivnik RJ, Petersen RC, Knopman D, Graff-Radford N, Parisi J, Dickson DW. DLB fluctuations: specific features that reliably differentiate DLB from AD and normal aging. Neurology. 2004;62:181–87. [DOI] [PubMed] [Google Scholar]
  • 276.O’Brien JT, Firbank MJ, Mosimann UP, Burn DJ, McKeith IG. Change in perfusion, hallucinations and fluctuations in consciousness in dementia with Lewy bodies. Psychiatry Res. 2005;139:79–88. [DOI] [PubMed] [Google Scholar]
  • 277.Schumacher J, Peraza LR, Firbank M, Thomas AJ, Kaiser M, Gallagher P, O’Brien JT, Blamire AM, Taylor JP. Dysfunctional brain dynamics and their origin in Lewy body dementia. Brain. 2019;142:1767–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Chabran E, Noblet V, Loureiro de Sousa P, Demuynck C, Philippi N, Mutter C, Anthony P, Martin-Hunyadi C, Cretin B, Blanc F. Changes in gray matter volume and functional connectivity in dementia with Lewy bodies compared to Alzheimer’s disease and normal aging: implications for fluctuations. Alzheimers Res Ther. 2020;12:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Taylor JP, Colloby SJ, McKeith IG, O’Brien JT. Covariant perfusion patterns provide clues to the origin of cognitive fluctuations and attentional dysfunction in dementia with Lewy bodies. Int PsychogeriAtr. 2013;25:1917–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Lowther ER, O’Brien JT, Firbank MJ, Blamire AM. Lewy body compared with Alzheimer dementia is associated with decreased functional connectivity in resting state networks. Psychiatry Res. 2014;223:192–201. [DOI] [PubMed] [Google Scholar]
  • 281.Morbelli S, Chincarini A, Brendel M, Rominger A, Bruffaerts R, Vandenberghe R, Kramberger MG, Trost M, Garibotto V, Nicastro N, et al. Metabolic patterns across core features in dementia with Lewy bodies. Ann Neurol. 2019;85:715–25. [DOI] [PubMed] [Google Scholar]
  • 282.Matar E, Ehgoetz Martens KA, Phillips JR, Wainstein G, Halliday GM, Lewis SJG, Shine JM. Dynamic network impairments underlie cognitive fluctuations in Lewy body dementia. NPJ Parkinsons Dis. 2022;8:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Franciotti R, Falasca NW, Bonanni L, Anzellotti F, Maruotti V, Comani S, Thomas A, Tartaro A, Taylor JP, Onofrj M. Default network is not hypoactive in dementia with fluctuating cognition: an Alzheimer disease/dementia with Lewy bodies comparison. Neurobiol Aging. 2013;34:1148–58. [DOI] [PubMed] [Google Scholar]
  • 284.Peraza LR, Kaiser M, Firbank M, Graziadio S, Bonanni L, Onofrj M, Colloby SJ, Blamire A, O’Brien J, Taylor JP. fMRI resting state networks and their association with cognitive fluctuations in dementia with Lewy bodies. neuroimage. Clin. 2014;4:558–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Matar E, Shine JM, Halliday GM, Lewis SJG. Cognitive fluctuations in Lewy body dementia: towards a pathophysiological framework. Brain. 2020;143:31–46. [DOI] [PubMed] [Google Scholar]
  • 286.Habich A, Wahlund LO, Westman E, Dierks T, Ferreira D. (Dis-)connected dots in dementia with Lewy bodies-a systematic review of connectivity studies. Mov Disord. 2023;38:4–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Boeve BF, Silber MH, Saper CB, Ferman TJ, Dickson DW, Parisi JE, Benarroch EE, Ahlskog JE, Smith GE, Caselli RC, et al. Pathophysiology of REM sleep behaviour disorder and relevance to neurodegenerative disease. Brain. 2007;130:2770–88. [DOI] [PubMed] [Google Scholar]
  • 288.Arrigoni E, Chen MC, Fuller PM. The anatomical, cellular and synaptic basis of motor atonia during rapid eye movement sleep. J Physiol. 2016;594:5391–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.García-Lorenzo D, Longo-Dos Santos C, Ewenczyk C, Leu-Semenescu S, Gallea C, Quattrocchi G, Pita Lobo P, Poupon C, Benali H, Arnulf I, et al. The coeruleus/subcoeruleus complex in rapid eye movement sleep behaviour disorders in Parkinson’s disease. Brain. 2013;136:2120–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Ehrminger M, Latimier A, Pyatigorskaya N, Garcia-Lorenzo D, Leu-Semenescu S, Vidailhet M, Lehericy S, Arnulf I. The coeruleus/subcoeruleus complex in idiopathic rapid eye movement sleep behaviour disorder. Brain. 2016;139:1180–88. [DOI] [PubMed] [Google Scholar]
  • 291.García-Gomar MG, Videnovic A, Singh K, Stauder M, Lewis LD, Wald LL, Rosen BR, Bianciardi M. Disruption of brainstem structural connectivity in REM sleep behavior disorder using 7 Tesla magnetic resonance imaging. Mov Disord. 2022;37:847–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Boeve BF, Silber MH, Ferman TJ, Lucas JA, Parisi JE. Association of REM sleep behavior disorder and neurodegenerative disease may reflect an underlying synucleinopathy. Mov Disord. 2001;16:622–30. [DOI] [PubMed] [Google Scholar]
  • 293.Ferman TJ, Boeve BF, Smith GE, Lin SC, Silber MH, Pedraza O, Wszolek Z, Graff-Radford NR, Uitti R, Van Gerpen J, et al. Inclusion of RBD improves the diagnostic classification of dementia with Lewy bodies. Neurology. 2011;77:875–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Boeve BF, Silber MH, Ferman TJ, Lin SC, Benarroch EE, Schmeichel AM, Ahlskog JE, Caselli RJ, Jacobson S, Sabbagh M, et al. Clinicopathologic correlations in 172 cases of rapid eye movement sleep behavior disorder with or without a coexisting neurologic disorder. Sleep Med. 2013;14:754–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Postuma RB, Adler CH, Dugger BN, Hentz JG, Shill HA, Driver-Dunckley E, Sabbagh MN, Jacobson SA, Belden CM, Sue LI, et al. REM sleep behavior disorder and neuropathology in Parkinson’s disease. Mov Disord. 2015;30:1413–17. [DOI] [PubMed] [Google Scholar]
  • 296.Leclair-Visonneau L, Clairembault T, Coron E, Le Dily S, Vavasseur F, Dalichampt M, Péréon Y, Neunlist M, Derkinderen P. REM sleep behavior disorder is related to enteric neuropathology in Parkinson disease. Neurology. 2017;89:1612–18. [DOI] [PubMed] [Google Scholar]
  • 297.Schmeichel AM, Buchhalter LC, Low PA, Parisi JE, Boeve BW, Sandroni P, Benarroch EE. Mesopontine cholinergic neuron involvement in Lewy body dementia and multiple system atrophy. Neurology. 2008;70:368–73. [DOI] [PubMed] [Google Scholar]
  • 298.Dugger BN, Murray ME, Boeve BF, Parisi JE, Benarroch EE, Ferman TJ, Dickson DW. Neuropathological analysis of brainstem cholinergic and catecholaminergic nuclei in relation to rapid eye movement (REM) sleep behaviour disorder. Neuropathol Appl NeuroBiol. 2012;38:142–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Grace KP, Horner RL. Evaluating the evidence surrounding pontine cholinergic involvement in REM Sleep generation. Front Neurol. 2015;6:190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Okkels N, Grothe MJ, Taylor JP, Hasselbalch SG, Fedorova TD, Knudsen K, van der Zee S, van Laar T, Bohnen NI, Borghammer P, Horsager J. Cholinergic changes in Lewy body disease: implications for presentation, progression and subtypes. Brain. 2024;147:2308–24. [DOI] [PubMed] [Google Scholar]
  • 301.Lu J, Sherman D, Devor M, Saper CB. A putative flip-flop switch for control of REM sleep. Nature. 2006;441:589–94. [DOI] [PubMed] [Google Scholar]
  • 302.Clément O, Sapin E, Bérod A, Fort P, Luppi PH. Evidence that neurons of the sublaterodorsal tegmental nucleus triggering paradoxical (REM) sleep are glutamatergic. Sleep. 2011;34:419–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Cox J, Pinto L, Dan Y. Calcium imaging of sleep-wake related neuronal activity in the dorsal pons. Nat Commun. 2016;7:10763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Boucetta S, Salimi A, Dadar M, Jones BE, Collins DL, Dang-Vu TT. Structural brain alterations associated with rapid eye movement Sleep behavior disorder in Parkinson’s disease. Sci Rep. 2016;6:26782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Torontali ZA, Fraigne JJ, Sanghera P, Horner R, Peever J. The sublaterodorsal tegmental nucleus functions to couple brain state and motor activity during REM sleep and wakefulness. Curr Biol. 2019;29:3803–13.e 3805. [DOI] [PubMed] [Google Scholar]
  • 306.Del Tredici K, Rüb, De Vos RA, Bohl JR, Braak H. Where does Parkinson disease pathology begin in the brain? J Neuropathol Exp Neurol. 2002;61:413–26. [DOI] [PubMed] [Google Scholar]
  • 307.Seidel K, Mahlke J, Siswanto S, Krüger R, Heinsen H, Auburger G, Bouzrou M, Grinberg LT, Wicht H, Korf HW, et al. The brainstem pathologies of Parkinson’s disease and dementia with Lewy bodies. Brain Pathol. 2015;25:121–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Taguchi K, Watanabe Y, Tsujimura A, Tanaka M. Brain region-dependent differential expression of alpha-synuclein. The J Comp Neurol. 2016;524:1236–58. [DOI] [PubMed] [Google Scholar]
  • 309.Feng H, Wen SY, Qiao QC, Pang YJ, Wang SY, Li HY, Cai J, Zhang KX, Chen J, Hu ZA, et al. Orexin signaling modulates synchronized excitation in the sublaterodorsal tegmental nucleus to stabilize REM sleep. Nat Commun. 2020;11:3661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Vetrivelan R, Bandaru SS. Neural control of REM sleep and motor atonia: current perspectives. Curr Neurol Neurosci Rep. 2023;23:907–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Kasanuki K, Iseki E, Kondo D, Fujishiro H, Minegishi M, Sato K, Katsuse O, Hino H, Kosaka K, Arai H. Neuropathological investigation of hypocretin expression in brains of dementia with Lewy bodies. Neurosci Lett. 2014;569:68–73. [DOI] [PubMed] [Google Scholar]
  • 312.Ferman TJ, Smith GE, Dickson DW, Graff-Radford NR, Lin SC, Wszolek Z, Van Gerpen JA, Uitti R, Knopman DS, Petersen RC, et al. Abnormal daytime sleepiness in dementia with Lewy bodies compared to Alzheimer’s disease using the multiple sleep latency test. Alzheimers Res Ther. 2014;6:76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Abbott RD, Ross GW, Duda JE, Shin C, Uyehara-Lock JH, Masaki KH, Launer LJ, White LR, Tanner CM, Petrovitch H. Excessive daytime sleepiness and topographic expansion of Lewy pathology. Neurology. 2019 1432;93:e1425–e. [DOI] [PMC free article] [PubMed]
  • 314.Mizutani M, Sano T, Ohira M, Takao M. Neuropathological studies of serotonergic and noradrenergic systems in Lewy body disease patients with delusion or depression. Psychiatry Clin Neurosci. 2022;76:459–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.Devanand DP, Lee S, Huey ED, Goldberg TE. Associations between neuropsychiatric symptoms and neuropathological diagnoses of Alzheimer disease and related dementias. JAMA Psychiarty. 2022;79:359–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Kasanuki K, Josephs KA, Ferman TJ, Murray ME, Koga S, Konno T, Sakae N, Parks A, Uitti RJ, Van Gerpen JA, et al. Diffuse Lewy body disease manifesting as corticobasal syndrome: a rare form of Lewy body disease. Neurology. 2018;91:e268–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Buciuc M, Whitwell JL, Kasanuki K, Graff-Radford J, Machulda MM, Duffy JR, Strand EA, Lowe VJ, Graff-Radford NR, Rush BK, et al. Lewy body disease is a contributor to logopenic progressive aphasia phenotype. Ann Neurol. 2021;89:520–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Braak H, Del Tredici K, Bratzke H, Hamm-Clement J, Sandmann-Keil D, Rüb U. Staging of the intracerebral inclusion body pathology associated with idiopathic Parkinson’s disease (preclinical and clinical stages). J Neurol. 2002;249(Suppl, 3):Iii/1–5. [DOI] [PubMed] [Google Scholar]
  • 319.Braak H, Rüb U, Gai WP. Del Tredici K. Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J Neural Transm (Vienna). 2003;110:517–36. [DOI] [PubMed] [Google Scholar]
  • 320.Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, Lashley T, Quinn NP, Rehncrona S, Björklund A, et al. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nat Med. 2008;14:501–03. [DOI] [PubMed] [Google Scholar]
  • 321.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;14:504–06. [DOI] [PubMed] [Google Scholar]
  • 322.Kordower JH, Chu Y, Hauser RA, Olanow CW, Freeman TB. Transplanted dopaminergic neurons develop PD pathologic changes: a second case report. Mov Disord. 2008;23:2303–06. [DOI] [PubMed] [Google Scholar]
  • 323.Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P, Trojanowski JQ, Lee VM. Pathological α-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science. 2012;338:949–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Masuda-Suzukake M, Nonaka T, Hosokawa M, Oikawa T, Arai T, Akiyama H, Mann DM, Hasegawa M. Prion-like spreading of pathological α-synuclein in brain. Brain. 2013;136:1128–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.Recasens A, Dehay B, Bové J, Carballo-Carbajal I, Dovero S, Pérez-Villalba A, Fernagut PO, Blesa J, Parent A, Perier C, 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. [DOI] [PubMed] [Google Scholar]
  • 326.Shimozawa A, Ono M, Takahara D, Tarutani A, Imura S, Masuda-Suzukake M, Higuchi M, Yanai K, Hisanaga SI, Hasegawa M. Propagation of pathological α-synuclein in marmoset brain. Acta Neuropathol Commun. 2017;5:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.Kim S, Kwon SH, Kam TI, Panicker N, Karuppagounder SS, Lee S, Lee JH, Kim WR, Kook M, Foss CA, et al. Transneuronal propagation of pathologic α-synuclein from the gut to the brain models Parkinson’s disease. Neuron. 2019;103:627–41.e627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Van Den Berge N, Ferreira N, Mikkelsen TW, Alstrup AKO, Tamgüney, Karlsson P, Terkelsen AJ, Nyengaard JR, Jensen PH, Borghammer P. Ageing promotes pathological alpha-synuclein propagation and autonomic dysfunction in wild-type rats. Brain. 2021;144:1853–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329.Rey NL, Steiner JA, Maroof N, Luk KC, Madaj Z, Trojanowski JQ, Lee VM, Brundin P. Widespread transneuronal propagation of α-synucleinopathy triggered in olfactory bulb mimics prodromal Parkinson’s disease. J Exp Med. 2016;213:1759–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Rey NL, George S, Steiner JA, Madaj Z, Luk KC, Trojanowski JQ, Lee VM, Brundin P. Spread of aggregates after olfactory bulb injection of α-synuclein fibrils is associated with early neuronal loss and is reduced long term. Acta Neuropathol. 2018;135:65–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331.Uemura N, Ueda J, Yoshihara T, Ikuno M, Uemura MT, Yamakado H, Asano M, Trojanowski JQ, Takahashi R. α-synuclein spread from olfactory bulb causes hyposmia, anxiety, and memory loss in BAC-SNCA mice. Mov Disord. 2021;36:2036–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Sawamura M, Onoe H, Tsukada H, Isa K, Yamakado H, Okuda S, Ikuno M, Hatanaka Y, Murayama S, Uemura N, et al. Lewy body disease Primate Model with α-Synuclein propagation from the olfactory bulb. Mov Disord. 2022;37:2033–44. [DOI] [PubMed] [Google Scholar]
  • 333.Jellinger KA. Is Braak staging valid for all types of Parkinson’s disease? J Neural Transm (Vienna). 2019;126:423–31. [DOI] [PubMed] [Google Scholar]
  • 334.Raunio A, Kaivola K, Tuimala J, Kero M, Oinas M, Polvikoski T, Paetau A, Tienari PJ, Myllykangas L. Lewy-related pathology exhibits two anatomically and genetically distinct progression patterns: a population-based study of finns aged 85. Acta Neuropathol. 2019;138:771–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335.Qualman SJ, Haupt HM, Yang P, Hamilton SR. Esophageal Lewy bodies associated with ganglion cell loss in achalasia. Similarity to Parkinson’s disease. Gastroenterology. 1984;87:848–56. [PubMed] [Google Scholar]
  • 336.Wakabayashi K, Takahashi H, Takeda S, Ohama E, Ikuta F. Parkinson’s disease: the presence of Lewy bodies in Auerbach’s and Meissner’s plexuses. Acta Neuropathol. 1988;76:217–21. [DOI] [PubMed] [Google Scholar]
  • 337.Svensson E, Horváth-Puhó E, Thomsen RW, Djurhuus JC, Pedersen L, Borghammer P, Sørensen HT. Does vagotomy reduce the risk of Parkinson’s disease: the authors reply. Ann Neurol. 2015;78:1012–13. [DOI] [PubMed] [Google Scholar]
  • 338.Liu B, Fang F, Pedersen NL, Tillander A, Ludvigsson JF, Ekbom A, Svenningsson P, Chen H, Wirdefeldt K. Vagotomy and Parkinson disease: a Swedish register-based matched-cohort study. Neurology. 2017;88:1996–2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Shannon KM, Keshavarzian A, Dodiya HB, Jakate S, Kordower JH. Is alpha-synuclein in the colon a biomarker for premotor Parkinson’s disease? Evidence from 3 cases. Mov Disord. 2012;27:716–19. [DOI] [PubMed] [Google Scholar]
  • 340.Hilton D, Stephens M, Kirk L, Edwards P, Potter R, Zajicek J, Broughton E, Hagan H, Carroll C. Accumulation of α-synuclein in the bowel of patients in the pre-clinical phase of Parkinson’s disease. Acta Neuropathol. 2014;127:235–41. [DOI] [PubMed] [Google Scholar]
  • 341.Stokholm MG, Danielsen EH, Hamilton-Dutoit SJ, Borghammer P. Pathological α-synuclein in gastrointestinal tissues from prodromal Parkinson disease patients. Ann Neurol. 2016;79:940–49. [DOI] [PubMed] [Google Scholar]
  • 342.Beck G, Hori Y, Hayashi Y, Morii E, Takehara T, Mochizuki H. Detection of phosphorylated alpha-synuclein in the muscularis propria of the gastrointestinal tract is a sensitive predictor for Parkinson’s disease. Parkinsons Dis. 2020;4687530. [DOI] [PMC free article] [PubMed]
  • 343.Beach TG, Adler CH, Sue LI, Serrano G, Shill HA, Walker DG, Lue L, Roher AE, Dugger BN, Maarouf C, et al. Arizona study of aging and neurodegenerative disorders and brain and body donation program. Neuropathology. 2015;35:354–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344.Adler CH, Beach TG. Neuropathological basis of nonmotor manifestations of Parkinson’s disease. Mov Disord. 2016;31:1114–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345.Beach TG, Adler CH, Sue LI, Shill HA, Driver-Dunckley E, Mehta SH, Intorcia AJ, Glass MJ, Walker JE, Arce R, 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 Parkinsons Dis. 2021;11:1833–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Sengoku R, Saito Y, Ikemura M, Hatsuta H, Sakiyama Y, Kanemaru K, Arai T, Sawabe M, Tanaka N, Mochizuki H, et al. Incidence and extent of Lewy body-related alpha-synucleinopathy in aging human olfactory bulb. J Neuropathol Exp Neurol. 2008;67:1072–83. [DOI] [PubMed] [Google Scholar]
  • 347.Miki Y, Mori F, Wakabayashi K, Kuroda N, Orimo S. Incidental Lewy body disease restricted to the heart and stellate ganglia. Mov Disord. 2009;24:2299–301. [DOI] [PubMed] [Google Scholar]
  • 348.Hawkes CH, Del Tredici K, Braak H. Parkinson’s disease: a dual-hit hypothesis. Neuropathol Appl Neurobiol. 2007;33:599–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Hawkes CH, Del Tredici K, Braak H. Parkinson’s disease: the dual hit theory revisited. Ann N Y Acad Sci. 2009, 1170;615–22. [DOI] [PubMed]
  • 350.Del Tredici K, Braak H. Review: sporadic Parkinson’s disease: development and distribution of α-synuclein pathology. Neuropathol Appl Neurobiol. 2016;42:33–50. [DOI] [PubMed] [Google Scholar]
  • 351.Borghammer P, Van Den Berge N. Brain-first versus gut-first Parkinson’s disease: a hypothesis. J Parkinsons Dis. 2019;9:S281–s295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Borghammer P. The α-Synuclein origin and connectome Model (SOC Model) of Parkinson’s disease: explaining motor asymmetry, non-motor phenotypes, and cognitive decline. J Parkinsons Dis. 2021;11:455–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Borghammer P, Horsager J, Andersen K, Van Den Berge N, Raunio A, Murayama S, Parkkinen L, Myllykangas L. Neuropathological evidence of body-first vs. brain-first Lewy body disease. Neurobiol Dis. 2021;161:105557. [DOI] [PubMed] [Google Scholar]
  • 354.Borghammer P, Just MK, Horsager J, Skjærbæk C, Raunio A, Kok EH, Savola S, Murayama S, Saito Y, Myllykangas L, Van Den Berge N. A postmortem study suggests a revision of the dual-hit hypothesis of Parkinson’s disease. NPJ Parkinsons Dis. 2022;8:166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355.Borghammer P. The brain-first vs. body-first model of Parkinson’s disease with comparison to alternative models. J Neural Transm (Vienna). 2023;130:737–53. [DOI] [PubMed] [Google Scholar]
  • 356.Uchihara T, Giasson BI. Propagation of alpha-synuclein pathology: hypotheses, discoveries, and yet unresolved questions from experimental and human brain studies. Acta Neuropathol. 2016;131:49–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Uchihara T. An order in Lewy body disorders: retrograde degeneration in hyperbranching axons as a fundamental structural template accounting for focal/multifocal Lewy body disease. Neuropathology. 2017;37:129–49. [DOI] [PubMed] [Google Scholar]
  • 358.Engelender S, Isacson O. The threshold theory for Parkinson’s disease. Trends Neurosci. 2017;40:4–14. [DOI] [PubMed] [Google Scholar]
  • 359.Surmeier DJ, Obeso JA, Halliday GM. Parkinson’s disease is not simply a prion disorder. J Neurosci. 2017;37:9799–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360.Gonzalez-Rodriguez P, Zampese E, Surmeier DJ. Selective neuronal vulnerability in Parkinson’s disease. Prog Brain Res. 2020;252:61–89. [DOI] [PubMed] [Google Scholar]
  • 361.Wakabayashi K. Where and how alpha-synuclein pathology spreads in Parkinson’s disease. Neuropathology. 2020;40:415–25. [DOI] [PubMed] [Google Scholar]
  • 362.Angot E, Steiner JA, Hansen C, Li JY, Brundin P. Are synucleinopathies prion-like disorders? The Lancet Neurol. 2010;9:1128–38. [DOI] [PubMed] [Google Scholar]
  • 363.Vargas JY, Grudina C, Zurzolo C. The prion-like spreading of α-synuclein: from in vitro to in vivo models of Parkinson’s disease. Ageing Res Rev. 2019;50:89–101. [DOI] [PubMed] [Google Scholar]
  • 364.Park H, Kam TI, Dawson VL, Dawson TM. α-Synuclein pathology as a target in neurodegenerative diseases. Nat Rev Neurol. 2025;21:32–47. [DOI] [PubMed] [Google Scholar]
  • 365.Uemura N. Fibril-seeded animal models of synucleinopathies: pathological mechanisms, disease modeling, and therapeutic implications. Neurosci Res. 2025;216:104905. [DOI] [PubMed] [Google Scholar]
  • 366.Luk KC, Song C, O’Brien P, Stieber A, Branch JR, Brunden KR, Trojanowski JQ, Lee VM. Exogenous alpha-synuclein fibrils seed the formation of Lewy body-like intracellular inclusions in cultured cells. Proc Natl Acad Sci USA. 2009;106:20051–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367.Danzer KM, Krebs SK, Wolff M, Birk G, Hengerer B. Seeding induced by alpha-synuclein oligomers provides evidence for spreading of alpha-synuclein pathology. J NeuroChem. 2009;111:192–203. [DOI] [PubMed] [Google Scholar]
  • 368.Aulić S, Le TT, Moda F, Abounit S, Corvaglia S, Casalis L, Gustincich S, Zurzolo C, Tagliavini F, Legname G. Defined α-synuclein prion-like molecular assemblies spreading in cell culture. BMC Neurosci. 2014;15:69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L, Spencer B, Masliah E, Lee SJ. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci USA. 2009;106:13010–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.Luk KC, Kehm VM, Zhang B, O’Brien P, Trojanowski JQ, Lee VM. Intracerebral inoculation of pathological α-synuclein initiates a rapidly progressive neurodegenerative α-synucleinopathy in mice. J Exp Med. 2012;209:975–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 371.Yamasaki TR, Holmes BB, Furman JL, Dhavale DD, Su BW, Song ES, Cairns NJ, Kotzbauer PT, Diamond MI. Parkinson’s disease and multiple system atrophy have distinct α-synuclein seed characteristics. J Biol Chem. 2019;294:1045–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Ayers JI, Lee J, Monteiro O, Woerman AL, Lazar AA, Condello C, Paras NA, Prusiner SB. Different α-synuclein prion strains cause dementia with Lewy bodies and multiple system atrophy. Proc Natl Acad Sci USA. 2022;119. [DOI] [PMC free article] [PubMed]
  • 373.Lau A, So RWL, Lau HHC, Sang JC, Ruiz-Riquelme A, Fleck SC, Stuart E, Menon S, Visanji NP, Meisl G, et al. α-Synuclein strains target distinct brain regions and cell types. Nat Neurosci. 2020;23:21–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Lloyd GM, Sorrentino ZA, Quintin S, Gorion KM, Bell BM, Paterno G, Long B, Prokop S, Giasson BI. Unique seeding profiles and prion-like propagation of synucleinopathies are highly dependent on the host in human α-synuclein transgenic mice. Acta Neuropathol. 2022;143:663–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 375.Zhang J, Han J, Shi Z, Zhang J, Zhou Z, Liu J, Yang G, Sun Y, Gu P, Zhao P, et al. The characteristic of nonmotor symptoms with different phenotypes and onsets in multiple system atrophy patients. J Clin Neurosci. 2023;109:1–5. [DOI] [PubMed] [Google Scholar]
  • 376.Prusiner SB, Woerman AL, Mordes DA, Watts JC, Rampersaud R, Berry DB, Patel S, Oehler A, Lowe JK, Kravitz SN, et al. Evidence for α-synuclein prions causing multiple system atrophy in humans with parkinsonism. Proc Natl Acad Sci USA. 2015;112:E5308–5317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 377.der Perren A V, Gelders G, Fenyi A, Bousset L, Brito F, Peelaerts W, den Haute C V, Gentleman S, Melki R, Baekelandt V. The structural differences between patient-derived α-synuclein strains dictate characteristics of Parkinson’s disease, multiple system atrophy and dementia with Lewy bodies. Acta Neuropathol. 2020;139:977–1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 378.Young AL, Marinescu RV, Oxtoby NP, Bocchetta M, Yong K, Firth NC, Cash DM, Thomas DL, Dick KM, Cardoso J, et al. Uncovering the heterogeneity and temporal complexity of neurodegenerative diseases with subtype and stage inference. Nat Commun. 2018;9:4273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 379.Young AL, Vogel JW, Aksman LM, Wijeratne PA, Eshaghi A, Oxtoby NP, Williams SCR, Alexander DC. Ordinal SuStaIn: subtype and stage inference for clinical scores, visual ratings, and other ordinal data. Front Artif Intell. 2021;4:613261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 380.Vogel JW, Young AL, Oxtoby NP, Smith R, Ossenkoppele R, Strandberg OT, La Joie R, Aksman LM, Grothe MJ, Iturria-Medina Y, et al. Four distinct trajectories of tau deposition identified in Alzheimer’s disease. Nat Med. 2021;27:871–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 381.Young AL, Bocchetta M, Russell LL, Convery RS, Peakman G, Todd E, Cash DM, Greaves CV, van Swieten J, Jiskoot L, et al. Characterizing the clinical features and atrophy patterns of MAPT-related frontotemporal dementia with disease progression modeling. Neurology. 2021;97:e941–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 382.Saito Y, Kamagata K, Wijeratne PA, Andica C, Uchida W, Takabayashi K, Fujita S, Akashi T, Wada A, Shimoji K, et al. Temporal progression patterns of brain atrophy in corticobasal syndrome and progressive supranuclear palsy revealed by subtype and stage inference (SuStain). Front Neurol. 2022;13:814768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 383.Collij LE, Salvadó G, Wottschel V, Mastenbroek SE, Schoenmakers P, Heeman F, Aksman L, Wink AM, Berckel BNM, van de Flier WM, et al. Spatial-temporal patterns of β-amyloid accumulation: a subtype and stage inference model analysis. Neurology. 2022 1703;98:e1692–e. [DOI] [PMC free article] [PubMed]
  • 384.Scotton WJ, Bocchetta M, Todd E, Cash DM, Oxtoby N, VandeVrede L, Heuer H, Alexander DC, Rowe JB, Morris HR, et al. A data-driven model of brain volume changes in progressive supranuclear palsy. Brain Commun. 2022;4:fcac098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 385.Scotton WJ, Shand C, Todd E, Bocchetta M, Cash DM, VandeVrede L, Heuer H, Young AL, Oxtoby N, Alexander DC, et al. Uncovering spatiotemporal patterns of atrophy in progressive supranuclear palsy using unsupervised machine learning. Brain Commun. 2023;5:fcad048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 386.Hong J, Lu J, Liu F, Wang M, Li X, Clement C, Lopes L, Brendel M, Rominger A, Yen TC, et al. Uncovering distinct progression patterns of tau deposition in progressive supranuclear palsy using [(18)F]Florzolotau PET imaging and subtype/stage inference algorithm. EBioMedicine. 2023;97:104835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 387.Zhou C, Wang L, Cheng W, Lv J, Guan X, Guo T, Wu J, Zhang W, Gao T, Liu X, et al. Two distinct trajectories of clinical and neurodegeneration events in Parkinson’s disease. NPJ Parkinsons Dis. 2023;9:111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 388.Sakato Y, Shima A, Terada Y, Takeda K, Sakamaki-Tsukita H, Nishida A, Yoshimura K, Wada I, Furukawa K, Kambe D, et al. Delineating three distinct spatiotemporal patterns of brain atrophy in Parkinson’s disease. Brain. 2024;147:3702–13. [DOI] [PubMed] [Google Scholar]
  • 389.Satoh R, Sekiya H, Ali F, Clark HM, Utianski RL, Duffy JR, Machulda MM, Dickson DW, Josephs KA, Whitwell JL. Uncovering image-driven subtypes with distinct pathology and clinical course in autopsy-confirmed four repeat Tauopathies. Ann Neurol. 2025. [DOI] [PMC free article] [PubMed]
  • 390.Young AL, Vogel JW, Robinson JL, McMillan CT, Ossenkoppele R, Wolk DA, Irwin DJ, Elman L, Grossman M, Lee VMY, et al. Data-driven neuropathological staging and subtyping of TDP-43 proteinopathies. Brain. 2023;146:2975–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 391.Mastenbroek SE, Vogel JW, Collij LE, Serrano GE, Tremblay C, Young AL, Arce RA, Shill HA, Driver-Dunckley ED, Mehta SH, et al. Disease progression modelling reveals heterogeneity in trajectories of Lewy-type α-synuclein pathology. Nat Commun. 2024;15:5133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 392.Otero-Jimenez M, Wojewska MJ, Binding LP, Jogaudaite S, Gray-Rodriguez S, Young AL, Gentleman S, Alegre-Abarrategui J. Neuropathological stages of neuronal, astrocytic and oligodendrocytic alpha-synuclein pathology in Parkinson’s disease. Acta Neuropathol Commun. 2025;13:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 393.Andersen KB, Krishnamurthy A, Just MK, Van Den Berge N, Skjærbæk C, Horsager J, Knudsen K, Vogel JW, Toledo JB, Attems J, et al. Sympathetic and parasympathetic subtypes of body-first Lewy body disease observed in postmortem tissue from prediagnostic individuals. Nat Neurosci. 2025;28:925–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 394.Nguyen PH, Ramamoorthy A, Sahoo BR, Zheng J, Faller P, Straub JE, Dominguez L, Shea JE, Dokholyan NV, De Simone A, et al. Amyloid oligomers: a joint Experimental/Computational perspective on Alzheimer’s disease, Parkinson’s disease, type II diabetes, and amyotrophic lateral sclerosis. Chem Rev. 2021;121:2545–647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 395.Winner B, Jappelli R, Maji SK, Desplats PA, Boyer L, Aigner S, Hetzer C, Loher T, Vilar M, Campioni S, et al. In vivo demonstration that alpha-synuclein oligomers are toxic. Proc Natl Acad Sci USA. 2011;108:4194–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 396.Cremades N, Cohen SI, Deas E, Abramov AY, Chen AY, Orte A, Sandal M, Clarke RW, Dunne P, Aprile FA, et al. Direct observation of the interconversion of normal and toxic forms of α-synuclein. Cell. 2012;149:1048–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397.Volles MJ, Lee SJ, Rochet JC, Shtilerman MD, Ding TT, Kessler JC, Lansbury PT Jr. Vesicle permeabilization by protofibrillar alpha-synuclein: implications for the pathogenesis and treatment of Parkinson’s disease. Biochemistry. 2001;40:7812–19. [DOI] [PubMed] [Google Scholar]
  • 398.Danzer KM, Haasen D, Karow AR, Moussaud S, Habeck M, Giese A, Kretzschmar H, Hengerer B, Kostka M. Different species of alpha-synuclein oligomers induce calcium influx and seeding. J Neurosci. 2007;27:9220–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 399.Chen SW, Drakulic S, Deas E, Ouberai M, Aprile FA, Arranz R, Ness S, Roodveldt C, Guilliams T, De-Genst EJ, et al. Structural characterization of toxic oligomers that are kinetically trapped during α-synuclein fibril formation. Proc Natl Acad Sci USA. 2015;112:E1994–2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 400.Angelova PR, Ludtmann MH, Horrocks MH, Negoda A, Cremades N, Klenerman D, Dobson CM, Wood NW, Pavlov EV, Gandhi S, Abramov AY. Ca2+ is a key factor in α-synuclein-induced neurotoxicity. J Cell Sci. 2016;129:1792–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401.Kaufmann TJ, Harrison PM, Richardson MJ, Pinheiro TJ, Wall MJ. Intracellular soluble α-synuclein oligomers reduce pyramidal cell excitability. J Physiol. 2016;594:2751–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402.Chaudhary H, Iyer A, Subramaniam V, Claessens MM. α-Synuclein oligomers stabilize pre-existing defects in supported bilayers and propagate membrane damage in a fractal-like pattern. Langmuir. 2016;32:11827–36. [DOI] [PubMed] [Google Scholar]
  • 403.Fusco G, Chen SW, Williamson PTF, Cascella R, Perni M, Jarvis JA, Cecchi C, Vendruscolo M, Chiti F, Cremades N, et al. Structural basis of membrane disruption and cellular toxicity by α-synuclein oligomers. Science. 2017;358:1440–43. [DOI] [PubMed] [Google Scholar]
  • 404.Vivoli Vega M, Cascella R, Chen SW, Fusco G, De Simone A, Dobson CM, Cecchi C, Chiti F. The toxicity of misfolded protein oligomers is Independent of their secondary structure. ACS Chem Biol. 2019;14:1593–600. [DOI] [PubMed] [Google Scholar]
  • 405.Cascella R, Chen SW, Bigi A, Camino JD, Xu CK, Dobson CM, Chiti F, Cremades N, Cecchi C. The release of toxic oligomers from α-synuclein fibrils induces dysfunction in neuronal cells. Nat Commun. 2021;12:1814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 406.Šneiderienė G, Czekalska MA, Xu CK, Jayaram AK, Krainer G, Arter WE, Peter QAE, Castellana-Cruz M, Saar KL, Levin A, et al. α-Synuclein oligomers displace monomeric α-Synuclein from lipid membranes. ACS Nano. 2024;18:17469–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 407.Chen L, Jin J, Davis J, Zhou Y, Wang Y, Liu J, Lockhart PJ, Zhang J. Oligomeric alpha-synuclein inhibits tubulin polymerization. Biochem Biophys Res Commun. 2007;356:548–53. [DOI] [PubMed] [Google Scholar]
  • 408.Chinta SJ, Mallajosyula JK, Rane A, Andersen JK. Mitochondrial α-synuclein accumulation impairs complex I function in dopaminergic neurons and results in increased mitophagy in vivo. Neurosci Lett. 2010;486:235–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 409.Luth ES, Stavrovskaya IG, Bartels T, Kristal BS, Selkoe DJ. Soluble, prefibrillar α-synuclein oligomers promote complex I-dependent, Ca2±induced mitochondrial dysfunction. J Biol Chem. 2014;289:21490–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 410.Plotegher N, Gratton E, Bubacco L. Number and brightness analysis of alpha-synuclein oligomerization and the associated mitochondrial morphology alterations in live cells. Biochim Biophys Acta. 2014;1840:2014–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 411.Lindström V, Gustafsson G, Sanders LH, Howlett EH, Sigvardson J, Kasrayan A, Ingelsson M, Bergström J, Erlandsson A. Extensive uptake of α-synuclein oligomers in astrocytes results in sustained intracellular deposits and mitochondrial damage. Mol Cell Neurosci. 2017;82:143–56. [DOI] [PubMed] [Google Scholar]
  • 412.Ludtmann MHR, Angelova PR, Horrocks MH, Choi ML, Rodrigues M, Baev AY, Berezhnov AV, Yao Z, Little D, Banushi B, et al. α-synuclein oligomers interact with ATP synthase and open the permeability transition pore in Parkinson’s disease. Nat Commun. 2018;9:2293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 413.Diógenes MJ, Dias RB, Rombo DM, Vicente Miranda H, Maiolino F, Guerreiro P, Näsström T, Franquelim HG, Oliveira LM, Castanho MA, et al. Extracellular alpha-synuclein oligomers modulate synaptic transmission and impair LTP via NMDA-receptor activation. J Neurosci. 2012;32:11750–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 414.Choi BK, Choi MG, Kim JY, Yang Y, Lai Y, Kweon DH, Lee NK, Shin YK. Large α-synuclein oligomers inhibit neuronal SNARE-mediated vesicle docking. Proc Natl Acad Sci USA. 2013;110:4087–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 415.Prots I, Veber V, Brey S, Campioni S, Buder K, Riek R, Böhm KJ, Winner B. α-synuclein oligomers impair neuronal microtubule-kinesin interplay. J Biol Chem. 2013;288:21742–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 416.Rockenstein E, Nuber S, Overk CR, Ubhi K, Mante M, Patrick C, Adame A, Trejo-Morales M, Gerez J, Picotti P, et al. Accumulation of oligomer-prone α-synuclein exacerbates synaptic and neuronal degeneration in vivo. Brain. 2014;137:1496–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 417.Ferreira DG, Temido-Ferreira M, Vicente Miranda H, Batalha VL, Coelho JE, É M S, Marques-Morgado I, Vaz SH, Rhee JS, Schmitz M, et al. α-synuclein interacts with PrP(C) to induce cognitive impairment through mGlur5 and NMDAR2B. Nat Neurosci. 2017;20:1569–79. [DOI] [PubMed] [Google Scholar]
  • 418.Plotegher N, Berti G, Ferrari E, Tessari I, Zanetti M, Lunelli L, Greggio E, Bisaglia M, Veronesi M, Girotto S, et al. DOPAL derived alpha-synuclein oligomers impair synaptic vesicles physiological function. Sci Rep. 2017;7:40699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 419.Durante V, de Iure A, Loffredo V, Vaikath N, De Risi M, Paciotti S, Quiroga-Varela A, Chiasserini D, Mellone M, Mazzocchetti P, et al. Alpha-synuclein targets GluN2A NMDA receptor subunit causing striatal synaptic dysfunction and visuospatial memory alteration. Brain. 2019;142:1365–85. [DOI] [PubMed] [Google Scholar]
  • 420.Jiang P, Gan M, Ebrahim AS, Lin WL, Melrose HL, Yen SH. ER stress response plays an important role in aggregation of α-synuclein. Mol Neurodegener. 2010;5:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 421.Colla E, Coune P, Liu Y, Pletnikova O, Troncoso JC, Iwatsubo T, Schneider BL, Lee MK. Endoplasmic reticulum stress is important for the manifestations of α-synucleinopathy in vivo. J Neurosci. 2012;32:3306–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 422.Colla E, Jensen PH, Pletnikova O, Troncoso JC, Glabe C, Lee MK. Accumulation of toxic α-synuclein oligomer within endoplasmic reticulum occurs in α-synucleinopathy in vivo. J Neurosci. 2012;32:3301–05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 423.Castillo-Carranza DL, Zhang Y, Guerrero-Muñoz MJ, Kayed R, Rincon-Limas DE, Fernandez-Funez P. Differential activation of the ER stress factor XBP1 by oligomeric assemblies. Neurochem Res. 2012;37:1707–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 424.Hughes CD, Choi ML, Ryten M, Hopkins L, Drews A, Botía JA, Iljina M, Rodrigues M, Gagliano SA, Gandhi S, et al. Picomolar concentrations of oligomeric alpha-synuclein sensitizes TLR4 to play an initiating role in Parkinson’s disease pathogenesis. Acta Neuropathol. 2019;137:103–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 425.Zhang W, Wang T, Pei Z, Miller DS, Wu X, Block ML, Wilson B, Zhang W, Zhou Y, Hong JS, Zhang J. Aggregated alpha-synuclein activates microglia: a process leading to disease progression in Parkinson’s disease. Faseb J. 2005;19:533–42. [DOI] [PubMed] [Google Scholar]
  • 426.Wilms H, Rosenstiel P, Romero-Ramos M, Arlt A, Schäfer H, Seegert D, Kahle PJ, Odoy S, Claasen JH, Holzknecht C, et al. Suppression of MAP kinases inhibits microglial activation and attenuates neuronal cell death induced by alpha-synuclein protofibrils. Int J Immunopathol Pharmacol. 2009;22:897–909. [DOI] [PubMed] [Google Scholar]
  • 427.Kim C, Ho DH, Suk JE, You S, Michael S, Kang J, Joong Lee S, Masliah E, Hwang D, Lee HJ, Lee SJ. Neuron-released oligomeric α-synuclein is an endogenous agonist of TLR2 for paracrine activation of microglia. Nat Commun. 2013;4:1562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 428.Yamaguchi H, Nishimura Y, Matsuse D, Sekiya H, Masaki K, Tanaka T, Saiga T, Harada M, Kira YI, Dickson DW, et al. A rapidly progressive multiple system atrophy-cerebellar variant model presenting marked glial reactions with inflammation and spreading of α-synuclein oligomers and phosphorylated α-synuclein aggregates. Brain Behav Immun. 2024. [DOI] [PubMed]
  • 429.Paleologou KE, Kragh CL, Mann DM, Salem SA, Al-Shami R, Allsop D, Hassan AH, Jensen PH, El-Agnaf OM. Detection of elevated levels of soluble alpha-synuclein oligomers in post-mortem brain extracts from patients with dementia with Lewy bodies. Brain. 2009;132:1093–101. [DOI] [PubMed] [Google Scholar]
  • 430.Tokuda T, Qureshi MM, Ardah MT, Varghese S, Shehab SA, Kasai T, Ishigami N, Tamaoka A, Nakagawa M, El-Agnaf OM. Detection of elevated levels of α-synuclein oligomers in CSF from patients with Parkinson disease. Neurology. 2010;75:1766–72. [DOI] [PubMed] [Google Scholar]
  • 431.Lassen LB, Gregersen E, Isager AK, Betzer C, Kofoed RH, Jensen PH. ELISA method to detect α-synuclein oligomers in cell and animal models. PLoS One. 2018;13:e0196056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 432.Saedi Z, Nikkhah M. A FRET-based aptasensor for the detection of α-synuclein oligomers as biomarkers of Parkinson’s disease. Anal Methods. 2022;14:2937–44. [DOI] [PubMed] [Google Scholar]
  • 433.Liu Y, Wang X, Campolo G, Teng X, Ying L, Edel JB, Ivanov AP. Single-molecule detection of α-synuclein oligomers in Parkinson’s disease patients using nanopores. ACS Nano. 2023;17:22999–3009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 434.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. [DOI] [PubMed] [Google Scholar]
  • 435.Söderberg O, Gullberg M, Jarvius M, Ridderstråle K, Leuchowius KJ, Jarvius J, Wester K, Hydbring P, Bahram F, Larsson LG, Landegren U. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat Methods. 2006;3:995–1000. [DOI] [PubMed] [Google Scholar]
  • 436.Roberts RF, Wade-Martins R, Alegre-Abarrategui J. Direct visualization of alpha-synuclein oligomers reveals previously undetected pathology in Parkinson’s disease brain. Brain. 2015;138:1642–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 437.Mazzetti S, Basellini MJ, Ferri V, Cassani E, Cereda E, Paolini M, Calogero AM, Bolliri C, De Leonardis M, Sacilotto G, et al. α-synuclein oligomers in skin biopsy of idiopathic and monozygotic twin patients with Parkinson’s disease. Brain. 2020;143:920–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 438.Behere A, Thörnqvist PO, Winberg S, Ingelsson M, Bergström J, Ekmark-Lewén S. Visualization of early oligomeric α-synuclein pathology and its impact on the dopaminergic system in the (thy-1)-h[A30P]α-syn transgenic mouse model. J Neurosci Res. 2021;99:2525–39. [DOI] [PubMed] [Google Scholar]
  • 439.Jensen NM, Fu Y, Betzer C, Li H, Elfarrash S, Shaib AH, Krah D, Vitic Z, Reimer L, Gram H, et al. MJF-14 proximity ligation assay detects early non-inclusion alpha-synuclein pathology with enhanced specificity and sensitivity. NPJ Parkinsons Dis. 2024;10:227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 440.Tanaka MT, Miki Y, Mori F, Kon T, Furukawa T, Shimoyama S, Tatara Y, Ozaki T, Bettencourt C, Warner TT, Wakabayashi K. Intranasal administration of trehalose reduces α-synuclein oligomers and accelerates α-synuclein aggregation. Brain Commun. 2024;6:fcae193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 441.Sekiya H, Franke L, Hashimoto Y, Takata M, Nishida K, Futamura N, Hasegawa K, Kowa H, Ross OA, McLean PJ, et al. Widespread distribution of α-synuclein oligomers in LRRK2-related Parkinson’s disease. Acta Neuropathol. 2025;149:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 442.Jensen NM, Vitic Z, Antorini MR, Viftrup TB, Parkkinen L, Jensen PH. Abundant non-inclusion α-synuclein pathology in Lewy body-negative LRRK2-mutant cases. Acta Neuropathol. 2025;149:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 443.Bartels T, Choi JG. Selkoe DJ. α-Synuclein occurs physiologically as a helically folded tetramer that resists aggregation. Nature. 2011;477:107–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 444.Dettmer U, Newman AJ, von Saucken VE, Bartels T, Selkoe D. KTKEGV repeat motifs are key mediators of normal α-synuclein tetramerization: their mutation causes excess monomers and neurotoxicity. Proc Natl Acad Sci USA. 2015;112:9596–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 445.Fauvet B, Mbefo MK, Fares MB, Desobry C, Michael S, Ardah MT, Tsika E, Coune P, Prudent M, Lion N, et al. α-synuclein in central nervous system and from erythrocytes, mammalian cells, and Escherichia coli exists predominantly as disordered monomer. J Biol Chem. 2012;287:15345–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 446.Miki Y, Tanji K, Shinnai K, Tanaka MT, Altay F, Foti SC, Strand C, Sasaki T, Kon T, Shimoyama S, et al. Pathological substrate of memory impairment in multiple system atrophy. Neuropathol Appl Neurobiol. 2022;e12844. [DOI] [PubMed]
  • 447.Sekiya H, Tsuji A, Hashimoto Y, Takata M, Koga S, Nishida K, Futamura N, Kawamoto M, Kohara N, Dickson DW, et al. Discrepancy between distribution of alpha-synuclein oligomers and Lewy-related pathology in Parkinson’s disease. Acta Neuropathol Commun. 2022;10:133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 448.Park MJ, Cheon SM, Bae HR, Kim SH, Kim JW. Elevated levels of α-synuclein oligomer in the cerebrospinal fluid of drug-naïve patients with Parkinson’s disease. J Clin Neurol. 2011;7:215–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 449.Parnetti L, Chiasserini D, Persichetti E, Eusebi P, Varghese S, Qureshi MM, Dardis A, Deganuto M, De Carlo C, Castrioto A, et al. Cerebrospinal fluid lysosomal enzymes and alpha-synuclein in Parkinson’s disease. Mov Disord. 2014;29:1019–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 450.Majbour NK, Vaikath NN, van Dijk KD, Ardah MT, Varghese S, Vesterager LB, Montezinho LP, Poole S, Safieh-Garabedian B, Tokuda T, et al. Oligomeric and phosphorylated alpha-synuclein as potential CSF biomarkers for Parkinson’s disease. Mol Neurodegener. 2016;11:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 451.Majbour NK, Abdi IY, Dakna M, Wicke T, Lang E, Ali Moussa HY, Thomas MA, Trenkwalder C, Safieh-Garabedian B, Tokuda T, et al. Cerebrospinal α-synuclein oligomers reflect disease motor severity in DeNoPa longitudinal cohort. Mov Disord. 2021;36:2048–56. [DOI] [PubMed] [Google Scholar]
  • 452.Blömeke L, Pils M, Kraemer-Schulien V, Dybala A, Schaffrath A, Kulawik A, Rehn F, Cousin A, Nischwitz V, Willbold J, et al. Quantitative detection of α-Synuclein and tau oligomers and other aggregates by digital single particle counting. NPJ Parkinsons Dis. 2022;8:68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 453.Hansson O, Hall S, Ohrfelt A, Zetterberg H, Blennow K, Minthon L, Nägga K, Londos E, Varghese S, Majbour NK, et al. Levels of cerebrospinal fluid α-synuclein oligomers are increased in Parkinson’s disease with dementia and dementia with Lewy bodies compared to Alzheimer’s disease. Alzheimers Res Ther. 2014;6:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 454.van Steenoven I, Majbour NK, Vaikath NN, Berendse HW, van der Flier Wm, van de Berg Wdj, Teunissen CE, Lemstra AW. El-Agnaf OMA. α-synuclein species as potential cerebrospinal fluid biomarkers for dementia with Lewy bodies. Mov Disord. 2018;33:1724–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 455.Sekiya H, Franke L, Ono D, DeTure M, Ross OA, Murray ME, McLean PJ, Ferman TJ, Dickson DW. Impact of hippocampal α-synuclein oligomers on cognitive trajectory in patients with dementia with Lewy bodies. Alzheimers Dement. 2025;21:e70374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 456.Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL, Bohr VA. Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol. 2019;15:565–81. [DOI] [PubMed] [Google Scholar]
  • 457.Choi JY, Morris JC, Hsu CY. Aging and cerebrovascular disease. Neurol Clin. 1998;16:687–711. [DOI] [PubMed] [Google Scholar]
  • 458.Smith MZ, Nagy Z, Barnetson L, King EM, Esiri MM. Coexisting pathologies in the brain: influence of vascular disease and Parkinson’s disease on Alzheimer’s pathology in the hippocampus. Acta Neuropathol. 2000;100:87–94. [DOI] [PubMed] [Google Scholar]
  • 459.Spires-Jones TL, Attems J, Thal DR. Interactions of pathological proteins in neurodegenerative diseases. Acta Neuropathol. 2017;134:187–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 460.Robinson JL, Lee EB, Xie SX, Rennert L, Suh E, Bredenberg C, Caswell C, Van Deerlin VM, Yan N, Yousef A, et al. Neurodegenerative disease concomitant proteinopathies are prevalent, age-related and APOE4-associated. Brain. 2018;141:2181–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 461.DeTure MA, Dickson DW. The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegener. 2019;14:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 462.Jecmenica Lukic M, Kurz C, Respondek G, Grau-Rivera O, Compta Y, Gelpi E, Troakes C, van Swieten Jc, Giese A, Roeber S, et al. Copathology in progressive supranuclear palsy: does it matter? Mov Disord. 2020;35:984–93. [DOI] [PubMed] [Google Scholar]
  • 463.Sekiya H, Koga S, Murakami A, DeTure M, Ross OA, Uitti RJ, Cheshire WP, Wszolek ZK, Dickson DW. Frequency of comorbid pathologies and their clinical impact in multiple system atrophy. Mov Disord. 2024;39:380–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 464.Braak H, Braak E. Neuropathological stageing of alzheimer-related changes. Acta Neuropathol. 1991;82:239–59. [DOI] [PubMed] [Google Scholar]
  • 465.Thal DR, Rüb U, Orantes M, Braak H. Phases of a beta-deposition in the human brain and its relevance for the development of AD. Neurology. 2002;58:1791–800. [DOI] [PubMed] [Google Scholar]
  • 466.Montine TJ, Phelps CH, Beach TG, Bigio EH, Cairns NJ, Dickson DW, Duyckaerts C, Frosch MP, Masliah E, Mirra SS, et al. National institute on aging-Alzheimer’s association guidelines for the neuropathologic assessment of Alzheimer’s disease: a practical approach. Acta Neuropathol. 2012;123:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 467.Polvikoski T, Sulkava R, Myllykangas L, Notkola IL, Niinistö L, Verkkoniemi A, Kainulainen K, Kontula K, Pérez-Tur J, Hardy J, Haltia M. Prevalence of Alzheimer’s disease in very elderly people: a prospective neuropathological study. Neurology. 2001;56:1690–96. [DOI] [PubMed] [Google Scholar]
  • 468.White L, Petrovitch H, Hardman J, Nelson J, Davis DG, Ross GW, Masaki K, Launer L, Markesbery WR. Cerebrovascular pathology and dementia in autopsied Honolulu-Asia aging study participants. Ann N Y Acad Sci. 2002;977:9–23. [DOI] [PubMed] [Google Scholar]
  • 469.Grodstein F, Leurgans SE, Capuano AW, Schneider JA, Bennett DA. Trends in postmortem neurodegenerative and cerebrovascular neuropathologies over 25 years. JAMA Neurol. 2023;80:370–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 470.Hyman BT, Trojanowski JQ. Editorial on consensus recommendations for the postmortem diagnosis of Alzheimer disease from the National Institute on Aging and the Reagan Institute working group on diagnostic criteria for the neuropathological assessment of Alzheimer disease. J Neuropathol & Exp Neurol. 1997;56:1095. [DOI] [PubMed] [Google Scholar]
  • 471.Jellinger KA, Attems J. Prevalence and impact of vascular and Alzheimer pathologies in Lewy body disease. Acta Neuropathol. 2008;115:427–36. [DOI] [PubMed] [Google Scholar]
  • 472.Nelson PT, Kryscio RJ, Jicha GA, Abner EL, Schmitt FA, Xu LO, Cooper G, Smith CD, Markesbery WR. Relative preservation of MMSE scores in autopsy-proven dementia with Lewy bodies. Neurology. 2009;73:1127–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 473.Kotzbauer PT, Cairns NJ, Campbell MC, Willis AW, Racette BA, Tabbal SD, Perlmutter JS. Pathologic accumulation of α-synuclein and Aβ in Parkinson disease patients with dementia. Arch Neurol. 2012;69:1326–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 474.Colom-Cadena M, Grau-Rivera O, Planellas L, Cerquera C, Morenas E, Helgueta S, Muñoz L, Kulisevsky J, Martí MJ, Tolosa E, et al. Regional overlap of pathologies in Lewy body disorders. J Neuropathol Exp Neurol. 2017;76:216–24. [DOI] [PubMed] [Google Scholar]
  • 475.McAleese KE, Walker L, Erskine D, Thomas AJ, McKeith IG, Attems J. TDP-43 pathology in Alzheimer’s disease, dementia with Lewy bodies and ageing. Brain Pathol. 2017;27:472–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 476.Tunold JA, Tan MMX, Koga S, Geut H, Rozemuller AJM, Valentino R, Sekiya H, Martin NB, Heckman MG, Bras J, et al. Lysosomal polygenic risk is associated with the severity of neuropathology in Lewy body disease. Brain. 2023. [DOI] [PMC free article] [PubMed]
  • 477.The National Institute on Aging and the Reagan Institute Working Group on Diagnostic Criteria for the Neuropathological Assessment of Alzheimer’s Disease. Consensus recommendations for the postmortem diagnosis of Alzheimer’s disease. Neurobiol Aging. 1997;18:S1–2. [PubMed] [Google Scholar]
  • 478.Chau E, Kim JR. α-synuclein-assisted oligomerization of β-amyloid (1-42). Arch Biochem Biophys. 2022;717:109120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 479.Candreva J, Chau E, Rice ME, Kim JR. Interactions between soluble species of β-amyloid and α-Synuclein promote oligomerization while inhibiting fibrillization. Biochemistry. 2020;59:425–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 480.Vadukul DM, Papp M, Thrush RJ, Wang J, Jin Y, Arosio P, Aprile FA. α-Synuclein aggregation is triggered by oligomeric amyloid-β 42 via heterogeneous primary nucleation. J Am Chem Soc. 2023;145:18276–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 481.Ono K, Takahashi R, Ikeda T, Yamada M. Cross-seeding effects of amyloid β-protein and α-synuclein. J NeuroChem. 2012;122:883–90. [DOI] [PubMed] [Google Scholar]
  • 482.Dasari AKR, Kayed R, Wi S, Lim KH. Tau interacts with the C-Terminal region of α-Synuclein, promoting formation of toxic aggregates with distinct molecular conformations. Biochemistry. 2019;58:2814–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 483.Lu J, Zhang S, Ma X, Jia C, Liu Z, Huang C, Liu C, Li D. Structural basis of the interplay between α-synuclein and tau in regulating pathological amyloid aggregation. J Biol Chem. 2020;295:7470–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 484.Torres-Garcia L, Jm PD, Brandi E, Haikal C, Mudannayake JM, Brás IC, Gerhardt E, Li W, Svanbergsson A, Outeiro TF, et al. Monitoring the interactions between alpha-synuclein and Tau in vitro and in vivo using bimolecular fluorescence complementation. Sci Rep. 2022;12:2987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 485.Fischer AL, Schmitz M, Thom T, Zafar S, Younas N, da Silva Correia S, da Silva Correia A, Eyyuboglu SC, Zerr I. Alpha-synuclein demonstrates varying binding affinities with different tau isoforms. J NeuroChem. 2025;169:e70053. [DOI] [PubMed] [Google Scholar]
  • 486.Badiola N, de Oliveira RM, Herrera F, Guardia-Laguarta C, Gonçalves SA, Pera M, Suárez-Calvet M, Clarimon J, Outeiro TF, Lleó A. Tau enhances α-synuclein aggregation and toxicity in cellular models of synucleinopathy. PLoS One. 2011;6:e26609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 487.Oikawa T, Nonaka T, Terada M, Tamaoka A, Hisanaga S, Hasegawa M. α-Synuclein fibrils exhibit gain of toxic function, promoting tau aggregation and inhibiting microtubule assembly. J Biol Chem. 2016;291:15046–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 488.Castillo-Carranza DL, Guerrero-Muñoz MJ, Sengupta U, Gerson JE, Kayed R. α-Synuclein oligomers induce a unique toxic Tau Strain. Biol Psychiatry. 2018;84:499–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 489.Gracia P, Polanco D, Tarancón-Díez J, Serra I, Bracci M, Oroz J, Laurents DV, García I, Cremades N. Molecular mechanism for the synchronized electrostatic coacervation and co-aggregation of alpha-synuclein and tau. Nat Commun. 2022;13:4586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 490.Sun C, Zhou K, Pt D, Li C, Lee VMY, Zhou ZH, Peng C, Jiang L. Structural basis of a distinct α-synuclein strain that promotes tau inclusion in neurons. J Biol Chem. 2025;301:108351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 491.Wei H, Masurkar AV, Razavian N. On gaps of clinical diagnosis of dementia subtypes: a study of Alzheimer’s disease and Lewy body disease. Front Aging Neurosci. 2023;15:1149036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 492.Sabbagh MN, Adler CH, Lahti TJ, Connor DJ, Vedders L, Peterson LK, Caviness JN, Shill HA, Sue LI, Ziabreva I, et al. Parkinson disease with dementia: comparing patients with and without Alzheimer pathology. Alzheimer Dis Assoc Disord. 2009;23:295–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 493.Compta Y, Parkkinen L, O’Sullivan SS, Vandrovcova J, Holton JL, Collins C, Lashley T, Kallis C, Williams DR, de Silva R, et al. Lewy- and alzheimer-type pathologies in Parkinson’s disease dementia: which is more important? Brain. 2011;134:1493–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 494.Toledo JB, Gopal P, Raible K, Irwin DJ, Brettschneider J, Sedor S, Waits K, Boluda S, Grossman M, Van Deerlin VM, et al. Pathological α-synuclein distribution in subjects with coincident Alzheimer’s and Lewy body pathology. Acta Neuropathol. 2016;131:393–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 495.Johnson DK, Morris JC, Galvin JE. Verbal and visuospatial deficits in dementia with Lewy bodies. Neurology. 2005;65:1232–38. [DOI] [PubMed] [Google Scholar]
  • 496.Coughlin D, Xie SX, Liang M, Williams A, Peterson C, Weintraub D, McMillan CT, Wolk DA, Akhtar RS, Hurtig HI, et al. Cognitive and pathological influences of tau pathology in Lewy body disorders. Ann Neurol. 2019;85:259–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 497.Boyle PA, Wang T, Yu L, Wilson RS, Dawe R, Arfanakis K, Schneider JA, Bennett DA. To what degree is late life cognitive decline driven by age-related neuropathologies? Brain. 2021;144:2166–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 498.Malek-Ahmadi M, Beach TG, Zamrini E, Adler CH, Sabbagh MN, Shill HA, Jacobson SA, Belden CM, Caselli RJ, Woodruff BK, et al. Faster cognitive decline in dementia due to Alzheimer disease with clinically undiagnosed Lewy body disease. PLoS One. 2019;14:e0217566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 499.de Boer Emj, Orie VK, Williams T, Baker MR, De Oliveira HM, Polvikoski T, Silsby M, Menon P, van den Bos M, Halliday GM, et al. TDP-43 proteinopathies: a new wave of neurodegenerative diseases. J Neurol Neurosurg Psychiatry. 2020;92:86–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 500.Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, Mori H, Mann D, Tsuchiya K, Yoshida M, Hashizume Y, Oda T. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun. 2006;351:602–11. [DOI] [PubMed] [Google Scholar]
  • 501.Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–33. [DOI] [PubMed] [Google Scholar]
  • 502.Keage HA, Hunter S, Matthews FE, Ince PG, Hodges J, Hokkanen SR, Highley JR, Dening T, Brayne C. TDP-43 pathology in the population: prevalence and associations with dementia and age. J Alzheimers Dis. 2014;42:641–50. [DOI] [PubMed] [Google Scholar]
  • 503.James BD, Wilson RS, Boyle PA, Trojanowski JQ, Bennett DA, Schneider JA. TDP-43 stage, mixed pathologies, and clinical Alzheimer’s-type dementia. Brain. 2016;139:2983–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 504.Flanagan ME, Cholerton B, Latimer CS, Hemmy LS, Edland SD, Montine KS, White LR, Montine TJ. TDP-43 neuropathologic associations in the Nun study and the Honolulu-asia Aging study. J Alzheimers Dis. 2018;66:1549–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 505.Mikhailenko E, Colangelo K, Tuimala J, Kero M, Savola S, Raunio A, Kok EH, Tanskanen M, Mäkelä M, Puttonen H, et al. Limbic-predominant age-related TDP-43 encephalopathy in the oldest old: a population-based study. Brain. 2025;148:154–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 506.Nakashima-Yasuda H, Uryu K, Robinson J, Xie SX, Hurtig H, Duda JE, Arnold SE, Siderowf A, Grossman M, Leverenz JB, et al. Co-morbidity of TDP-43 proteinopathy in Lewy body related diseases. Acta Neuropathol. 2007;114:221–29. [DOI] [PubMed] [Google Scholar]
  • 507.Higashi S, Iseki E, Yamamoto R, Minegishi M, Hino H, Fujisawa K, Togo T, Katsuse O, Uchikado H, Furukawa Y, et al. Concurrence of TDP-43, tau and alpha-synuclein pathology in brains of Alzheimer’s disease and dementia with Lewy bodies. Brain Res. 2007;1184:284–94. [DOI] [PubMed] [Google Scholar]
  • 508.Arai T, Mackenzie IR, Hasegawa M, Nonoka T, Niizato K, Tsuchiya K, Iritani S, Onaya M, Akiyama H. Phosphorylated TDP-43 in Alzheimer’s disease and dementia with Lewy bodies. Acta Neuropathol. 2009;117:125–36. [DOI] [PubMed] [Google Scholar]
  • 509.Meneses A, Koga S, O’Leary J, Dickson DW, Bu G, Zhao N. TDP-43 pathology in Alzheimer’s disease. Mol Neurodegener. 2021;16:84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 510.Hasegawa M, Arai T, Nonaka T, Kametani F, Yoshida M, Hashizume Y, Beach TG, Buratti E, Baralle F, Morita M, et al. Phosphorylated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Ann Neurol. 2008;64:60–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 511.Murakami A, Koga S, Sekiya H, Oskarsson B, Boylan K, Petrucelli L, Josephs KA, Dickson DW. Old age amyotrophic lateral sclerosis and limbic TDP-43 pathology. Brain Pathol. 2022;e13100. [DOI] [PMC free article] [PubMed]
  • 512.Jamerlan AM. An SSA. A microplate-based approach to map interactions between TDP-43 and α-synuclein. J Clin Med. 2022;11. [DOI] [PMC free article] [PubMed]
  • 513.Dhakal S, Wyant CE, George HE, Morgan SE, Rangachari V. Prion-like C-Terminal Domain of TDP-43 and α-Synuclein interact synergistically to generate neurotoxic hybrid fibrils. J Mol Biol. 2021;433:166953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 514.Dhakal S, Robang AS, Bhatt N, Puangmalai N, Fung L, Kayed R, Paravastu AK, Rangachari V. Distinct neurotoxic TDP-43 fibril polymorphs are generated by heterotypic interactions with α-synuclein. J Biol Chem. 2022;298:102498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 515.Buciuc M, Whitwell JL, Boeve BF, Ferman TJ, Graff-Radford J, Savica R, Kantarci K, Fields JA, Knopman DS, Petersen RC, et al. TDP-43 is associated with a reduced likelihood of rendering a clinical diagnosis of dementia with Lewy bodies in autopsy-confirmed cases of transitional/diffuse Lewy body disease. J Neurol. 2020;267:1444–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 516.Josephs KA, Whitwell JL, Weigand SD, Murray ME, Tosakulwong N, Liesinger AM, Petrucelli L, Senjem ML, Knopman DS, Boeve BF, et al. TDP-43 is a key player in the clinical features associated with Alzheimer’s disease. Acta Neuropathol. 2014;127:811–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 517.Uemura MT, Robinson JL, Cousins KAQ, Tropea TF, Kargilis DC, McBride JD, Suh E, Xie SX, Xu Y, Porta S, et al. Distinct characteristics of limbic-predominant age-related TDP-43 encephalopathy in Lewy body disease. Acta Neuropathol. 2022;143:15–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 518.O’Brien JT. Thomas A. Vascular dementia. Lancet. 2015;386:1698–706. [DOI] [PubMed] [Google Scholar]
  • 519.Inoue Y, Shue F, Bu G, Kanekiyo T. Pathophysiology and probable etiology of cerebral small vessel disease in vascular dementia and Alzheimer’s disease. Mol Neurodegener. 2023;18:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 520.Dupré N, Drieu A, Joutel A. Pathophysiology of cerebral small vessel disease: a journey through recent discoveries. J Clin Invest 2024;134. [DOI] [PMC free article] [PubMed]
  • 521.Isojima D, Togo T, Kosaka K, Fujishiro H, Akatsu H, Katsuse O, Iritani S, Matsumoto T, Hirayasu Y. Vascular complications in dementia with Lewy bodies: a postmortem study. Neuropathology. 2006;26:293–97. [DOI] [PubMed] [Google Scholar]
  • 522.Jellinger KA. Prevalence of cerebrovascular lesions in Parkinson’s disease. A postmortem study. Acta Neuropathol. 2003;105:415–19. [DOI] [PubMed] [Google Scholar]
  • 523.Ghebremedhin E, Rosenberger A, Rüb U, Vuksic M, Berhe T, Bickeböller H, de Vos RA, Thal DR, Deller T. Inverse relationship between cerebrovascular lesions and severity of Lewy body pathology in patients with Lewy body diseases. J Neuropathol Exp Neurol. 2010;69:442–48. [DOI] [PubMed] [Google Scholar]
  • 524.Thal DR, Ghebremedhin E, Orantes M, Wiestler OD. Vascular pathology in Alzheimer disease: correlation of cerebral amyloid angiopathy and arteriosclerosis/lipohyalinosis with cognitive decline. J Neuropathol Exp Neurol. 2003;62:1287–301. [DOI] [PubMed] [Google Scholar]
  • 525.Chen ZC, Gan J, Yang Y, Meng Q, Han J, Ji Y. The vascular risk factors and vascular neuropathology in subjects with autopsy-confirmed dementia with Lewy bodies. Int J Geriatr Psychiatry. 2022;37. [DOI] [PMC free article] [PubMed]
  • 526.Agrawal S, Leurgans SE, Nag S, Oveisgharan S, Barnes LL, Bennett DA, Buchman AS, Schneider JA. Effects of cerebrovascular and Lewy body pathology on parkinsonian signs in community-dwelling older adults. Neurology. 2023;101:e754–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 527.Hase Y, Polvikoski TM, Firbank MJ, Craggs LJL, Hawthorne E, Platten C, Stevenson W, Deramecourt V, Ballard C, Kenny RA, et al. Small vessel disease pathological changes in neurodegenerative and vascular dementias concomitant with autonomic dysfunction. Brain Pathol. 2020;30:191–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 528.Thal DR, Griffin WS, de Vos RA. Ghebremedhin E. Cerebral amyloid angiopathy and its relationship to Alzheimer’s disease. Acta Neuropathol. 2008;115:599–609. [DOI] [PubMed] [Google Scholar]
  • 529.Xuereb JH, Brayne C, Dufouil C, Gertz H, Wischik C, Harrington C, Mukaetova-Ladinska E, McGee MA, O’Sullivan A, O’Connor D, et al. Neuropathological findings in the very old. Results from the first 101 brains of a population-based longitudinal study of dementing disorders. Ann N Y Acad Sci. 2000;903:490–96. [DOI] [PubMed] [Google Scholar]
  • 530.Pfeifer LA, White LR, Ross GW, Petrovitch H, Launer LJ. Cerebral amyloid angiopathy and cognitive function: the HAAS autopsy study. Neurology. 2002;58:1629–34. [DOI] [PubMed] [Google Scholar]
  • 531.Boyle PA, Yu L, Nag S, Leurgans S, Wilson RS, Bennett DA, Schneider JA. Cerebral amyloid angiopathy and cognitive outcomes in community-based older persons. Neurology. 2015;85:1930–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 532.Mäkelä M, Paetau A, Polvikoski T, Myllykangas L, Tanskanen M. Capillary amyloid-β protein deposition in a population-based study (vantaa 85+). J Alzheimers Dis. 2016;49:149–57. [DOI] [PubMed] [Google Scholar]
  • 533.Revesz T, Ghiso J, Lashley T, Plant G, Rostagno A, Frangione B, Holton JL. Cerebral amyloid angiopathies: a pathologic, biochemical, and genetic view. J Neuropathol Exp Neurol. 2003;62:885–98. [DOI] [PubMed] [Google Scholar]
  • 534.Vonsattel JP, Myers RH, Hedley-Whyte ET, Ropper AH, Bird ED, Richardson EP Jr. Cerebral amyloid angiopathy without and with cerebral hemorrhages: a comparative histological study. Ann Neurol. 1991;30:637–49. [DOI] [PubMed] [Google Scholar]
  • 535.Thal DR, Ghebremedhin E, Rüb U, Yamaguchi H, Del Tredici K, Braak H. Two types of sporadic cerebral amyloid angiopathy. J Neuropathol Exp Neurol. 2002;61:282–93. [DOI] [PubMed] [Google Scholar]
  • 536.Love S, Chalmers K, Ince P, Esiri M, Attems J, Jellinger K, Yamada M, McCarron M, Minett T, Matthews F, et al. Development, appraisal, validation and implementation of a consensus protocol for the assessment of cerebral amyloid angiopathy in post-mortem brain tissue. Am J Neurodegener Dis. 2014;3:19–32. [PMC free article] [PubMed] [Google Scholar]
  • 537.Jellinger KA. Dementia with grains (argyrophilic grain disease). Brain Pathol. 1998;8:377–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 538.Ferrer I, Santpere G, van Leeuwen FW. Argyrophilic grain disease. Brain. 2008;131:1416–32. [DOI] [PubMed] [Google Scholar]
  • 539.Josephs KA, Whitwell JL, Parisi JE, Knopman DS, Boeve BF, Geda YE, Jack CR Jr., Petersen RC, Dickson DW. Argyrophilic grains: a distinct disease or an additive pathology? Neurobiol Aging. 2008;29:566–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 540.Yokota O, Miki T, Nakashima-Yasuda H, Ishizu H, Haraguchi T, Ikeda C, Hasegawa M, Miyashita A, Ikeuchi T, Nishikawa N, et al. Pure argyrophilic grain disease revisited: independent effects on limbic, neocortical, and striato-pallido-nigral degeneration and the development of dementia in a series with a low to moderate Braak stage. Acta Neuropathol Commun. 2024;12:121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 541.Tolnay M, Spillantini MG, Goedert M, Ulrich J, Langui D, Probst A. Argyrophilic grain disease: widespread hyperphosphorylation of tau protein in limbic neurons. Acta Neuropathol. 1997;93:477–84. [DOI] [PubMed] [Google Scholar]
  • 542.Martinez-Lage P, Munoz DG. Prevalence and disease associations of argyrophilic grains of Braak. J Neuropathol Exp Neurol. 1997;56:157–64. [DOI] [PubMed] [Google Scholar]
  • 543.Braak H, Braak E. Argyrophilic grain disease: frequency of occurrence in different age categories and neuropathological diagnostic criteria. J Neural Transm (Vienna). 1998;105:801–19. [DOI] [PubMed] [Google Scholar]
  • 544.Braak H, Braak E. Cortical and subcortical argyrophilic grains characterize a disease associated with adult onset dementia. Neuropathol Appl neurobiol. 1989;15:13–26. [DOI] [PubMed] [Google Scholar]
  • 545.Iseki E, Togo T, Suzuki K, Katsuse O, Marui W, de Silva R, Lees A, Yamamoto T, Kosaka K. Dementia with Lewy bodies from the perspective of tauopathy. Acta Neuropathol. 2003;105:265–70. [DOI] [PubMed] [Google Scholar]
  • 546.Sabbagh MN, Sandhu SS, Farlow MR, Vedders L, Shill HA, Caviness JN, Connor DJ, Sue L, Adler CH, Beach TG. Correlation of clinical features with argyrophilic grains at autopsy. Alzheimer Dis Assoc Disord. 2009;23:229–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 547.Homma T, Mochizuki Y, Takahashi K, Komori T. Medial temporal regional argyrophilic grain as a possible important factor affecting dementia in Parkinson’s disease. Neuropathology. 2015;35:441–51. [DOI] [PubMed] [Google Scholar]
  • 548.Saito Y, Ruberu NN, Sawabe M, Arai T, Tanaka N, Kakuta Y, Yamanouchi H, Murayama S. Staging of argyrophilic grains: an age-associated tauopathy. J Neuropathol Exp Neurol. 2004;63:911–18. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Data Availability Statement

Not applicable.


Articles from Molecular Neurodegeneration are provided here courtesy of BMC

RESOURCES