Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.299
Filtrer
1.
Mol Neurodegener ; 19(1): 59, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090623

RÉSUMÉ

BACKGROUND: Multiple lines of evidence support peripheral organs in the initiation or progression of Lewy body disease (LBD), a spectrum of neurodegenerative diagnoses that include Parkinson's Disease (PD) without or with dementia (PDD) and dementia with Lewy bodies (DLB). However, the potential contribution of the peripheral immune response to LBD remains unclear. This study aims to characterize peripheral immune responses unique to participants with LBD at single-cell resolution to highlight potential biomarkers and increase mechanistic understanding of LBD pathogenesis in humans. METHODS: In a case-control study, peripheral mononuclear cell (PBMC) samples from research participants were randomly sampled from multiple sites across the United States. The diagnosis groups comprise healthy controls (HC, n = 159), LBD (n = 110), Alzheimer's disease dementia (ADD, n = 97), other neurodegenerative disease controls (NDC, n = 19), and immune disease controls (IDC, n = 14). PBMCs were activated with three stimulants (LPS, IL-6, and IFNa) or remained at basal state, stained by 13 surface markers and 7 intracellular signal markers, and analyzed by flow cytometry, which generated 1,184 immune features after gating. RESULTS: The model classified LBD from HC with an AUROC of 0.87 ± 0.06 and AUPRC of 0.80 ± 0.06. Without retraining, the same model was able to distinguish LBD from ADD, NDC, and IDC. Model predictions were driven by pPLCγ2, p38, and pSTAT5 signals from specific cell populations under specific activation. The immune responses characteristic for LBD were not associated with other common medical conditions related to the risk of LBD or dementia, such as sleep disorders, hypertension, or diabetes. CONCLUSIONS AND RELEVANCE: Quantification of PBMC immune response from multisite research participants yielded a unique pattern for LBD compared to HC, multiple related neurodegenerative diseases, and autoimmune diseases thereby highlighting potential biomarkers and mechanisms of disease.


Sujet(s)
Agranulocytes , Maladie à corps de Lewy , Maladie de Parkinson , Humains , Maladie de Parkinson/immunologie , Maladie de Parkinson/métabolisme , Maladie à corps de Lewy/immunologie , Mâle , Femelle , Sujet âgé , Études cas-témoins , Agranulocytes/métabolisme , Agranulocytes/immunologie , Marqueurs biologiques/métabolisme , Adulte d'âge moyen , Études de cohortes , Sujet âgé de 80 ans ou plus , Corps de Lewy/anatomopathologie , Corps de Lewy/métabolisme , Analyse sur cellule unique/méthodes
2.
Acta Neuropathol ; 148(1): 18, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39141121

RÉSUMÉ

Lewy body disorders are heterogeneous neurological conditions defined by intracellular inclusions composed of misshapen α-synuclein protein aggregates. Although α-synuclein aggregates are only one component of inclusions and not strictly coupled to neurodegeneration, evidence suggests they seed the propagation of Lewy pathology within and across cells. Genetic mutations, genomic multiplications, and sequence polymorphisms of the gene encoding α-synuclein are also causally linked to Lewy body disease. In nonfamilial cases of Lewy body disease, the disease trigger remains unidentified but may range from industrial/agricultural toxicants and natural sources of poisons to microbial pathogens. Perhaps due to these peripheral exposures, Lewy inclusions appear at early disease stages in brain regions connected with cranial nerves I and X, which interface with inhaled and ingested environmental elements in the nasal or gastrointestinal cavities. Irrespective of its identity, a stealthy disease trigger most likely shifts soluble α-synuclein (directly or indirectly) into insoluble, cross-ß-sheet aggregates. Indeed, ß-sheet-rich self-replicating α-synuclein multimers reside in patient plasma, cerebrospinal fluid, and other tissues, and can be subjected to α-synuclein seed amplification assays. Thus, clinicians should be able to capitalize on α-synuclein seed amplification assays to stratify patients into potential responders versus non-responders in future clinical trials of α-synuclein targeted therapies. Here, we briefly review the current understanding of α-synuclein in Lewy body disease and speculate on pathophysiological processes underlying the potential transmission of α-synucleinopathy across the neuraxis.


Sujet(s)
Maladie à corps de Lewy , alpha-Synucléine , Humains , Maladie à corps de Lewy/anatomopathologie , Maladie à corps de Lewy/métabolisme , alpha-Synucléine/métabolisme , Animaux , Encéphale/anatomopathologie , Encéphale/métabolisme , Corps de Lewy/anatomopathologie , Corps de Lewy/métabolisme
3.
Rinsho Shinkeigaku ; 64(8): 557-563, 2024 Aug 27.
Article de Japonais | MEDLINE | ID: mdl-39069492

RÉSUMÉ

The patient was an 85-year-old man with a one-year history of difficulty reading kana. Neuropsychological evaluation revealed kana (phonogram)-selective reading impairment and kanji (ideogram)-dominant writing impairment. MRI revealed significant cerebral atrophy in the left occipital cortex, leading to the clinical diagnosis of posterior cortical atrophy (PCA). Cerebrospinal fluid amyloid ß1-42 levels were reduced, and amyloid PET showed accumulation in the posterior cingulate cortex, precuneus, and frontal lobe. In contrast, tau PET showed no accumulation in the atrophied brain areas. Episodes of REM sleep behavior disorder and decreased uptake on meta-iodobenzylguanidine (MIBG) myocardial scintigraphy suggested the involvement of Lewy body pathology. PCA with distinct laterality has been rarely reported, and |this is the first case to present Kana-selective reading impairment and Kanji-dominant writing impairment with neurodegenerative background.


Sujet(s)
Atrophie , Imagerie par résonance magnétique , Humains , Mâle , Sujet âgé de 80 ans ou plus , Tomographie par émission de positons , Dyslexie/étiologie , Cortex cérébral/anatomopathologie , Cortex cérébral/imagerie diagnostique , Marqueurs biologiques/liquide cérébrospinal , Peptides bêta-amyloïdes/métabolisme , Peptides bêta-amyloïdes/liquide cérébrospinal , Lobe occipital/anatomopathologie , Lobe occipital/imagerie diagnostique , Corps de Lewy/anatomopathologie , Trouble du comportement en sommeil paradoxal/étiologie , Trouble du comportement en sommeil paradoxal/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/diagnostic
4.
Clin Auton Res ; 34(3): 329-339, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38844644

RÉSUMÉ

PURPOSE: Neurogenic orthostatic hypotension (nOH) results from deficient reflexive delivery of norepinephrine to cardiovascular receptors in response to decreased cardiac venous return. Lewy body (LB) forms of nOH are characterized by low 18F-dopamine-derived radioactivity (a measure of cardiac noradrenergic deficiency), olfactory dysfunction by the University of Pennsylvania Smell Identification Test (UPSIT), and increased deposition of alpha-synuclein (α-syn) in dermal sympathetic noradrenergic nerves by the α-syn-tyrosine hydroxylase (TH) colocalization index. This observational, cross-sectional study explored whether combinations of these biomarkers specifically identify LB forms of nOH. METHODS: Clinical laboratory data were reviewed from patients referred for evaluation at the National Institutes of Health for chronic autonomic failure between 2011 and 2023. The cutoff value for low myocardial 18F-dopamine-derived radioactivity was 6000 nCi-kg/cc-mCi, for olfactory dysfunction an UPSIT score ≤ 28, and for an increased α-syn-TH colocalization index ≥ 1.57. RESULTS: A total of 44 patients (31 LB, 13 non-LB nOH) had data for all three biomarkers. Compared to the non-LB group, the LB nOH group had low myocardial 18F-dopamine-derived radioactivity, low UPSIT scores, and high α-syn-TH colocalization indexes (p < 0.0001 each). Combining the three biomarkers completely separated the groups. Cluster analysis identified two distinct groups (p < 0.0001) independently of the clinical diagnosis, with one cluster corresponding exactly to LB nOH. CONCLUSION: LB forms of nOH feature cardiac noradrenergic deficiency, olfactory dysfunction, and increased α-syn-TH colocalization in skin biopsies. Combining the data for these variables efficiently separates LB from non-LB nOH. Independently of the clinical diagnosis, this biomarker triad identifies a pathophysiologically distinct cluster of nOH patients.


Sujet(s)
Marqueurs biologiques , Hypotension orthostatique , Humains , Hypotension orthostatique/diagnostic , Hypotension orthostatique/physiopathologie , Mâle , Femelle , Sujet âgé , Marqueurs biologiques/analyse , Études transversales , Adulte d'âge moyen , alpha-Synucléine/métabolisme , Corps de Lewy/anatomopathologie , Dopamine/analogues et dérivés , Dopamine/métabolisme , Sujet âgé de 80 ans ou plus
5.
Neurodegener Dis ; 24(1): 6-15, 2024.
Article de Anglais | MEDLINE | ID: mdl-38861955

RÉSUMÉ

INTRODUCTION: Sleep disturbances have been associated with essential tremor (ET). However, their pathophysiological underpinnings remain unknown. In this exploratory study, we examined the association between subjective sleep disturbances and the presence of Lewy pathology (LP) on postmortem brain examination in ET cases. METHODS: Fifty-two ET cases enrolled in a prospective, longitudinal study were assessed over an average period of 42 months. Cases completed the Pittsburgh Sleep Quality Index (PSQI), which yields seven component scores (e.g., sleep quality, sleep latency). For each component score, we calculated the difference between the last score and the baseline score. Brains were harvested at death. Each had a complete neuropathological assessment, including extensive α-synuclein immunostaining. We examined the associations between baseline PSQI scores and the change in PSQI scores (last - first), and LP on postmortem brain examination. RESULTS: ET cases had a mean baseline age of 87.1 ± 4.8 years. LP was observed in 12 (23.1%) of 52 cases; in 7 of these 12, LP was observed in the locus coeruleus (LC). Change in time needed to fall asleep (last - first sleep latency component score) was associated with presence of LP on postmortem brain examination - greater increase in sleep latency was associated with higher odds of LP (odds ratio = 2.98, p = 0.02). The greatest increase in sleep latency was observed in cases with LP in the LC (p = 0.04). CONCLUSION: In ET cases, increases in sleep latency over time could be a marker of underlying LP, especially in the LC.


Sujet(s)
Encéphale , Tremblement essentiel , Troubles de la veille et du sommeil , Humains , Tremblement essentiel/anatomopathologie , Femelle , Mâle , Troubles de la veille et du sommeil/anatomopathologie , Troubles de la veille et du sommeil/épidémiologie , Sujet âgé de 80 ans ou plus , Études longitudinales , Sujet âgé , Encéphale/anatomopathologie , Études prospectives , Corps de Lewy/anatomopathologie , Études de cohortes , alpha-Synucléine/métabolisme
6.
Nat Commun ; 15(1): 5133, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879548

RÉSUMÉ

Lewy body (LB) diseases, characterized by the aggregation of misfolded α-synuclein proteins, exhibit notable clinical heterogeneity. This may be due to variations in accumulation patterns of LB neuropathology. Here we apply a data-driven disease progression model to regional neuropathological LB density scores from 814 brain donors with Lewy pathology. We describe three inferred trajectories of LB pathology that are characterized by differing clinicopathological presentation and longitudinal antemortem clinical progression. Most donors (81.9%) show earliest pathology in the olfactory bulb, followed by accumulation in either limbic (60.8%) or brainstem (21.1%) regions. The remaining donors (18.1%) initially exhibit abnormalities in brainstem regions. Early limbic pathology is associated with Alzheimer's disease-associated characteristics while early brainstem pathology is associated with progressive motor impairment and substantial LB pathology outside of the brain. Our data provides evidence for heterogeneity in the temporal spread of LB pathology, possibly explaining some of the clinical disparities observed in Lewy body disease.


Sujet(s)
Évolution de la maladie , Corps de Lewy , Maladie à corps de Lewy , alpha-Synucléine , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Humains , Mâle , Adulte d'âge moyen , alpha-Synucléine/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Encéphale/anatomopathologie , Encéphale/métabolisme , Tronc cérébral/anatomopathologie , Tronc cérébral/métabolisme , Corps de Lewy/anatomopathologie , Corps de Lewy/métabolisme , Maladie à corps de Lewy/anatomopathologie , Maladie à corps de Lewy/métabolisme , Bulbe olfactif/anatomopathologie , Bulbe olfactif/métabolisme
7.
J Neurochem ; 168(7): 1215-1236, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38693066

RÉSUMÉ

Parkinson's disease (PD) is a common neurodegenerative disorder that is affecting an increasing number of older adults. Although PD is mostly sporadic, genetic mutations have been found in cohorts of families with a history of familial PD (FPD). The first such mutation linked to FPD causes a point mutation (A53T) in α-synuclein (α-syn), a major component of Lewy bodies, which are a classical pathological hallmark of PD. These findings suggest that α-syn is an important contributor to the development of PD. In our previous study, we developed an adenoviral mouse model of PD and showed that the expression of wild-type (WT) α-syn or a mutant form with an increased propensity to aggregate, designated as WT-CL1 α-syn, could be used to study how α-syn aggregation contributes to PD. In this study, we established a transgenic mouse model that conditionally expresses WT or WT-CL1 α-syn in dopaminergic (DA) neurons and found that the expression of either WT or WT-CL1 α-syn was associated with an age-dependent degeneration of DA neurons and movement dysfunction. Using this model, we were able to monitor the process of α-syn aggregate formation and found a correlation between age and the number and sizes of α-syn aggregates formed. These results provide a potential mechanism by which age-dependent α-syn aggregation may lead to the formation of Lewy bodies in PD pathogenesis.


Sujet(s)
Vieillissement , Corps de Lewy , Souris transgéniques , Maladie de Parkinson , alpha-Synucléine , Animaux , Humains , Mâle , Souris , Vieillissement/génétique , Vieillissement/métabolisme , alpha-Synucléine/métabolisme , alpha-Synucléine/génétique , Modèles animaux de maladie humaine , Neurones dopaminergiques/métabolisme , Neurones dopaminergiques/anatomopathologie , Corps de Lewy/métabolisme , Corps de Lewy/anatomopathologie , Souris de lignée C57BL , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie
8.
Article de Anglais | MEDLINE | ID: mdl-38757945

RÉSUMÉ

BACKGROUND: Dementia results from multiple neuropathologies causing cognitive impairment sufficiently severe to affect functional status. However, these pathologies and functional impairment are common in persons without dementia. We examined the association of Alzheimer's disease (AD) and multiple other neuropathologies with instrumental and basic activities of daily living in persons with and without dementia. METHODS: Participants were 1 509 deceased from the Religious Orders Study or Rush Memory and Aging Project. Pathologic AD and 3 other AD indices were examined, in addition to 4 non-AD neurodegenerative pathologies: cerebral amyloid angiopathy (CAA), hippocampal sclerosis, TDP-43, and Lewy bodies, and 4 cerebrovascular pathologies: gross- and microinfarctions, athero- and arteriolosclerosis. Functional assessment included Lawton and Katz Index Instrumental and Basic Activities of Daily Living (IADL and BADL). Ordinal regression models adjusted for age, sex, and education were used to examine the association of neuropathologies with IADL and BADL. RESULTS: Alzheimer's disease and the other neuropathologies were associated with impaired IADL (all ps < .001) and with impaired BADL (ps < .01), except for atherosclerosis and CAA, which were not associated with BADL. The effects of most neuropathologies were largely affected by dementia. However, small effects on IADL remained for PHF-tau tangles after adjusting models for dementia. Direct effects of gross infarcts on IADL and BADL and of microinfarcts on BADL remained unchanged after adjusting the models for dementia. CONCLUSIONS: Alzheimer's disease and all other neuropathologies are strongly associated with functional disability. The association of most neuropathologies with disability was eliminated or attenuated by dementia, except for gross infarcts and microinfarcts.


Sujet(s)
Activités de la vie quotidienne , Maladie d'Alzheimer , Humains , Maladie d'Alzheimer/anatomopathologie , Femelle , Mâle , Sujet âgé de 80 ans ou plus , Sujet âgé , Démence , Angiopathie amyloïde cérébrale/anatomopathologie , Corps de Lewy/anatomopathologie , Hippocampe/anatomopathologie , Évaluation de l'invalidité
9.
Alzheimers Dement ; 20(8): 5114-5131, 2024 08.
Article de Anglais | MEDLINE | ID: mdl-38770829

RÉSUMÉ

INTRODUCTION: Alzheimer's disease (AD) pathology is defined by ß-amyloid (Aß) plaques and neurofibrillary tau, but Lewy bodies (LBs; 𝛼-synuclein aggregates) are a common co-pathology for which effective biomarkers are needed. METHODS: A validated α-synuclein Seed Amplification Assay (SAA) was used on recent cerebrospinal fluid (CSF) samples from 1638 Alzheimer's Disease Neuroimaging Initiative (ADNI) participants, 78 with LB-pathology confirmation at autopsy. We compared SAA outcomes with neuropathology, Aß and tau biomarkers, risk-factors, genetics, and cognitive trajectories. RESULTS: SAA showed 79% sensitivity and 97% specificity for LB pathology, with superior performance in identifying neocortical (100%) compared to limbic (57%) and amygdala-predominant (60%) LB-pathology. SAA+ rate was 22%, increasing with disease stage and age. Higher Aß burden but lower CSF p-tau181 associated with higher SAA+ rates, especially in dementia. SAA+ affected cognitive impairment in MCI and Early-AD who were already AD biomarker positive. DISCUSSION: SAA is a sensitive, specific marker for LB-pathology. Its increase in prevalence with age and AD stages, and its association with AD biomarkers, highlights the clinical importance of α-synuclein co-pathology in understanding AD's nature and progression. HIGHLIGHTS: SAA shows 79% sensitivity, 97% specificity for LB-pathology detection in AD. SAA positivity prevalence increases with disease stage and age. Higher Aß burden, lower CSF p-tau181 linked with higher SAA+ rates in dementia. SAA+ impacts cognitive impairment in early disease stages. Study underpins need for wider LB-pathology screening in AD treatment.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Marqueurs biologiques , Neuroimagerie , alpha-Synucléine , Protéines tau , Humains , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/liquide cérébrospinal , alpha-Synucléine/liquide cérébrospinal , Marqueurs biologiques/liquide cérébrospinal , Mâle , Femelle , Sujet âgé , Études transversales , Protéines tau/liquide cérébrospinal , Peptides bêta-amyloïdes/liquide cérébrospinal , Sujet âgé de 80 ans ou plus , Prévalence , Corps de Lewy/anatomopathologie , Cognition/physiologie , Sensibilité et spécificité , Encéphale/anatomopathologie , Encéphale/imagerie diagnostique , Dysfonctionnement cognitif/génétique , Dysfonctionnement cognitif/liquide cérébrospinal
10.
Acta Neuropathol Commun ; 12(1): 81, 2024 05 24.
Article de Anglais | MEDLINE | ID: mdl-38790074

RÉSUMÉ

Cerebrovascular and α-synuclein pathologies are frequently observed alongside Alzheimer disease (AD). The heterogeneity of AD necessitates comprehensive approaches to postmortem studies, including the representation of historically underrepresented ethnic groups. In this cohort study, we evaluated small vessel disease pathologies and α-synuclein deposits among Hispanic decedents (HD, n = 92) and non-Hispanic White decedents (NHWD, n = 184) from three Alzheimer's Disease Research Centers: Columbia University, University of California San Diego, and University of California Davis. The study included cases with a pathological diagnosis of Intermediate/High AD based on the National Institute on Aging- Alzheimer's Association (NIA-AA) and/or NIA-Reagan criteria. A 2:1 random comparison sample of NHWD was frequency-balanced and matched with HD by age and sex. An expert blinded to demographics and center origin evaluated arteriolosclerosis, cerebral amyloid angiopathy (CAA), and Lewy bodies/Lewy neurites (LBs/LNs) with a semi-quantitative approach using established criteria. There were many similarities and a few differences among groups. HD showed more severe Vonsattel grading of CAA in the cerebellum (p = 0.04), higher CAA density in the posterior hippocampus and cerebellum (ps = 0.01), and increased LBs/LNs density in the frontal (p = 0.01) and temporal cortices (p = 0.03), as determined by Wilcoxon's test. Ordinal logistic regression adjusting for age, sex, and center confirmed these findings except for LBs/LNs in the temporal cortex. Results indicate HD with AD exhibit greater CAA and α-synuclein burdens in select neuroanatomic regions when compared to age- and sex-matched NHWD with AD. These findings aid in the generalizability of concurrent arteriolosclerosis, CAA, and LBs/LNs topography and severity within the setting of pathologically confirmed AD, particularly in persons of Hispanic descent, showing many similarities and a few differences to those of NHW descent and providing insights into precision medicine approaches.


Sujet(s)
Maladie d'Alzheimer , Hispanique ou Latino , Corps de Lewy , , Humains , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/ethnologie , Femelle , Mâle , Sujet âgé , Sujet âgé de 80 ans ou plus , Études de cohortes , Corps de Lewy/anatomopathologie , Angiopathie amyloïde cérébrale/anatomopathologie , Angiopathie amyloïde cérébrale/ethnologie , alpha-Synucléine/métabolisme , Encéphale/anatomopathologie , Maladies des petits vaisseaux cérébraux/anatomopathologie , Maladies des petits vaisseaux cérébraux/ethnologie , Artériolosclérose/anatomopathologie
11.
Mov Disord Clin Pract ; 11(7): 874-878, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38757351

RÉSUMÉ

BACKGROUND: The G2019S leucine-rich repeat kinase 2 (LRRK2) gene mutation is an important and commonly found genetic determinant of Parkinson's disease (PD). The neuropathological findings associated with this mutation have thus far been varied but are most often associated with Lewy body (LB) pathology. OBJECTIVE: Describe a case of clinical Parkinson's disease with levodopa responsiveness found to have LRRK2 mutations and the absence of Lewy bodies. METHOD: We present an 89-year-old man with a 10-year history of slowly progressive parkinsonism suspected to be secondary to Parkinson's disease. RESULTS: Neuropathological evaluation revealed nigral degeneration without Lewy bodies or Lewy neurites, but there were frequent tau-immunopositive neurites and astrocytes in the putamen and substantia nigra, neocortical glial tau positive astrocytes associated with aging-related tau astrogliopathy (ARTAG), as well as neurofibrillary tangles, beta amyloid plaques, and amyloid angiopathy typical of advanced Alzheimer's disease. G2019S LRRK2 homozygous mutations were found. CONCLUSION: This case illustrates that levodopa-responsive clinical PD caused by G2019S LRRK2 mutations can occur without Lewy bodies.


Sujet(s)
Leucine-rich repeat serine-threonine protein kinase-2 , Corps de Lewy , Mutation , Maladie de Parkinson , Protein-Serine-Threonine Kinases , Humains , Leucine-rich repeat serine-threonine protein kinase-2/génétique , Leucine-rich repeat serine-threonine protein kinase-2/métabolisme , Mâle , Maladie de Parkinson/génétique , Maladie de Parkinson/anatomopathologie , Sujet âgé de 80 ans ou plus , Corps de Lewy/anatomopathologie , Corps de Lewy/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protéines tau/génétique , Protéines tau/métabolisme , Lévodopa/usage thérapeutique
12.
Neurochem Int ; 177: 105760, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38723900

RÉSUMÉ

Neurodegenerative diseases such as Parkinson's disease (PD) are characterized by the death of neurons in specific areas of the brain. One of the proteins that is involved in the pathogenesis of PD is α-synuclein (α-syn). α-Syn is a normal protein that is found in all neurons, but in PD, it misfolds and aggregates into toxic fibrils. These fibrils can then coalesce into pathological inclusions, such as Lewy bodies and Lewy neurites. The pathogenic pathway of PD is thought to involve a number of steps, including misfolding and aggregation of α-syn, mitochondrial dysfunction, protein clearance impairment, neuroinflammation and oxidative stress. A deeper insight into the structure of α-syn and its fibrils could aid in understanding the disease's etiology. The prion-like nature of α-syn is also an important area of research. Prions are misfolded proteins that can spread from cell to cell, causing other proteins to misfold as well. It is possible that α-syn may behave in a similar way, spreading from cell to cell and causing a cascade of misfolding and aggregation. Various post-translational alterations have also been observed to play a role in the pathogenesis of PD. These alterations can involve a variety of nuclear and extranuclear activities, and they can lead to the misfolding and aggregation of α-syn. A better understanding of the pathogenic pathway of PD could lead to the development of new therapies for the treatment of this disease.


Sujet(s)
Maladie de Parkinson , Pliage des protéines , alpha-Synucléine , alpha-Synucléine/métabolisme , Humains , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Animaux , Corps de Lewy/métabolisme , Corps de Lewy/anatomopathologie
13.
Alzheimers Dement ; 20(6): 4351-4365, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38666355

RÉSUMÉ

INTRODUCTION: Amyloid beta and tau pathology are the hallmarks of sporadic Alzheimer's disease (AD) and autosomal dominant AD (ADAD). However, Lewy body pathology (LBP) is found in ≈ 50% of AD and ADAD brains. METHODS: Using an α-synuclein seed amplification assay (SAA) in cerebrospinal fluid (CSF) from asymptomatic (n = 26) and symptomatic (n = 27) ADAD mutation carriers, including 12 with known neuropathology, we investigated the timing of occurrence and prevalence of SAA positive reactivity in ADAD in vivo. RESULTS: No asymptomatic participant and only 11% (3/27) of the symptomatic patients tested SAA positive. Neuropathology revealed LBP in 10/12 cases, primarily affecting the amygdala or the olfactory areas. In the latter group, only the individual with diffuse LBP reaching the neocortex showed α-synuclein seeding activity in CSF in vivo. DISCUSSION: Results suggest that in ADAD LBP occurs later than AD pathology and often as amygdala- or olfactory-predominant LBP, for which CSF α-synuclein SAA has low sensitivity. HIGHLIGHTS: Cerebrospinal fluid (CSF) real-time quaking-induced conversion (RT-QuIC) detects misfolded α-synuclein in ≈ 10% of symptomatic autosomal dominant Alzheimer's disease (ADAD) patients. CSF RT-QuIC does not detect α-synuclein seeding activity in asymptomatic mutation carriers. Lewy body pathology (LBP) in ADAD mainly occurs as olfactory only or amygdala-predominant variants. LBP develops late in the disease course in ADAD. CSF α-synuclein RT-QuIC has low sensitivity for focal, low-burden LBP.


Sujet(s)
Maladie d'Alzheimer , Corps de Lewy , alpha-Synucléine , Humains , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/liquide cérébrospinal , alpha-Synucléine/liquide cérébrospinal , alpha-Synucléine/génétique , Femelle , Mâle , Adulte d'âge moyen , Corps de Lewy/anatomopathologie , Sujet âgé , Mutation , Encéphale/anatomopathologie , Peptides bêta-amyloïdes/liquide cérébrospinal , Peptides bêta-amyloïdes/métabolisme , Évolution de la maladie
14.
Mov Disord ; 39(7): 1212-1217, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38597193

RÉSUMÉ

BACKGROUND: While preclinical studies have shown that alpha-synuclein can spread through cell-to-cell transmission whether it can be transmitted between humans is unknown. OBJECTIVES: The aim was to assess the presence of a synucleinopathy in autopsied conjugal couples. METHODS: Neuropathological findings in conjugal couples were categorized as Parkinson's disease (PD), dementia with Lewy bodies (DLB), Alzheimer's disease with Lewy bodies (ADLB), incidental Lewy body disease (ILBD), or no Lewy bodies. RESULTS: Ninety conjugal couples were included; the mean age of death was 88.3 years; 32 couples had no Lewy bodies; 42 couples had 1 spouse with a synucleinopathy: 10 PD, 3 DLB, 13 ADLB, and 16 ILBD; 16 couples had both spouses with a synucleinopathy: in 4 couples both spouses had PD, 1 couple had PD and DLB, 4 couples had PD and ADLB, 2 couples had PD and ILBD, 1 couple had DLB and ADLB, in 3 couples both had ADLB, and 1 couple had ADLB and ILBD. No couples had both spouses with ILBD. CONCLUSIONS: This large series of 90 autopsied conjugal couples found 16 conjugal couples with synucleinopathies, suggesting transmission of synucleinopathy between spouses is unlikely. © 2024 International Parkinson and Movement Disorder Society.


Sujet(s)
Maladie à corps de Lewy , Maladie de Parkinson , Synucléinopathies , Humains , Femelle , Mâle , Sujet âgé de 80 ans ou plus , Maladie de Parkinson/anatomopathologie , Maladie de Parkinson/métabolisme , Maladie à corps de Lewy/anatomopathologie , Maladie à corps de Lewy/métabolisme , Sujet âgé , Synucléinopathies/anatomopathologie , Autopsie , alpha-Synucléine/métabolisme , Conjoints , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Corps de Lewy/anatomopathologie
15.
Acta Neuropathol ; 147(1): 67, 2024 04 06.
Article de Anglais | MEDLINE | ID: mdl-38581586

RÉSUMÉ

Transcription factor EB (TFEB) is a master regulator of genes involved in the maintenance of autophagic and lysosomal homeostasis, processes which have been implicated in the pathogenesis of GBA-related and sporadic Parkinson's disease (PD), and dementia with Lewy bodies (DLB). TFEB activation results in its translocation from the cytosol to the nucleus. Here, we investigated TFEB subcellular localization and its relation to intracellular alpha-synuclein (aSyn) accumulation in post-mortem human brain of individuals with either incidental Lewy body disease (iLBD), GBA-related PD/DLB (GBA-PD/DLB) or sporadic PD/DLB (sPD/DLB), compared to control subjects. We analyzed nigral dopaminergic neurons using high-resolution confocal and stimulated emission depletion (STED) microscopy and semi-quantitatively scored the TFEB subcellular localization patterns. We observed reduced nuclear TFEB immunoreactivity in PD/DLB patients compared to controls, both in sporadic and GBA-related cases, as well as in iLBD cases. Nuclear depletion of TFEB was more pronounced in neurons with Ser129-phosphorylated (pSer129) aSyn accumulation in all groups. Importantly, we observed previously-unidentified TFEB-immunopositive perinuclear clusters in human dopaminergic neurons, which localized at the Golgi apparatus. These TFEB clusters were more frequently observed and more severe in iLBD, sPD/DLB and GBA-PD/DLB compared to controls, particularly in pSer129 aSyn-positive neurons, but also in neurons lacking detectable aSyn accumulation. In aSyn-negative cells, cytoplasmic TFEB clusters were more frequently observed in GBA-PD/DLB and iLBD patients, and correlated with reduced GBA enzymatic activity as well as increased Braak LB stage. Altered TFEB distribution was accompanied by a reduction in overall mRNA expression levels of selected TFEB-regulated genes, indicating a possible early dysfunction of lysosomal regulation. Overall, we observed cytoplasmic TFEB retention and accumulation at the Golgi in cells without apparent pSer129 aSyn accumulation in iLBD and PD/DLB patients. This suggests potential TFEB impairment at the early stages of cellular disease and underscores TFEB as a promising therapeutic target for synucleinopathies.


Sujet(s)
Maladie à corps de Lewy , Humains , alpha-Synucléine/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Encéphale/anatomopathologie , Neurones dopaminergiques/métabolisme , Corps de Lewy/anatomopathologie , Maladie à corps de Lewy/anatomopathologie
16.
J Neuroinflammation ; 21(1): 93, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38622654

RÉSUMÉ

The neuroinflammatory process in synucleinopathies of the aging population such as Parkinson's disease (PD) and dementia with Lewy bodies (DLB) involves microglial activation as well as infiltration of the CNS by T cells and natural killer T cells (NKTs). To evaluate the potential of targeting NKT cells to modulate neuroinflammation, we treated α-syn transgenic (tg) mice (e.g.: Thy1 promoter line 61) with an antibody against CD1d, which is a glycoprotein expressed in antigen presenting cells (APCs). CD1d-presented lipid antigens activate NKT cells through the interaction with T cell receptor in NKTs, resulting in the production of cytokines. Thus, we hypothesized that blocking the APC-NKT interaction with an anti-CD1d antibody might reduce neuroinflammation and neurodegeneration in models of DLB/PD. Treatment with the anti-CD1d antibody did not have effects on CD3 (T cells), slightly decreased CD4 and increased CD8 lymphocytes in the mice. Moreover, double labeling studies showed that compared to control (IgG) treated α-syn tg mice, treatment with anti-CD1d decreased numbers of CD3/interferon γ (IFN γ)-positive cells, consistent with NKTs. Further double labeling studies showed that CD1d-positive cells co-localized with the astrocytes marker GFAP and that anti-CD1d antibody reduced this effect. While in control α-syn tg mice CD3 positive cells were near astrocytes, this was modified by the treatment with the CD1d antibody. By qPCR, levels of IFN γ, CCL4, and interleukin-6 were increased in the IgG treated α-syn tg mice. Treatment with CD1d antibody blunted this cytokine response that was associated with reduced astrocytosis and microgliosis in the CNS of the α-syn tg mice treated with CD1d antibody. Flow cytometric analysis of immune cells in α-syn tg mice revealed that CD1d-tet + T cells were also increased in the spleen of α-syn tg mice, which treatment with the CD1d antibody reduced. Reduced neuroinflammation in the anti-CD1d-treated α-syn tg mice was associated with amelioration of neurodegenerative pathology. These results suggest that reducing infiltration of NKT cells with an antibody against CD1d might be a potential therapeutical approach for DLB/PD.


Sujet(s)
Maladie de Parkinson , alpha-Synucléine , Souris , Animaux , alpha-Synucléine/génétique , Corps de Lewy/anatomopathologie , Maladies neuro-inflammatoires , Maladie de Parkinson/anatomopathologie , Souris transgéniques , Immunothérapie/méthodes , Cytokines , Immunoglobuline G
17.
Nat Commun ; 15(1): 2750, 2024 Mar 29.
Article de Anglais | MEDLINE | ID: mdl-38553463

RÉSUMÉ

The defining feature of Parkinson disease (PD) and Lewy body dementia (LBD) is the accumulation of alpha-synuclein (Asyn) fibrils in Lewy bodies and Lewy neurites. Here we develop and validate a method to amplify Asyn fibrils extracted from LBD postmortem tissue samples and use solid state nuclear magnetic resonance (SSNMR) studies to determine atomic resolution structure. Amplified LBD Asyn fibrils comprise a mixture of single protofilament and two protofilament fibrils with very low twist. The protofilament fold is highly similar to the fold determined by a recent cryo-electron microscopy study for a minority population of twisted single protofilament fibrils extracted from LBD tissue. These results expand the structural characterization of LBD Asyn fibrils and approaches for studying disease mechanisms, imaging agents and therapeutics targeting Asyn.


Sujet(s)
Maladie à corps de Lewy , Maladie de Parkinson , Humains , alpha-Synucléine/composition chimique , Cryomicroscopie électronique , Corps de Lewy/anatomopathologie , Maladie à corps de Lewy/anatomopathologie , Maladie de Parkinson/anatomopathologie
18.
Parkinsonism Relat Disord ; 122: 106077, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38461037

RÉSUMÉ

These facts argue against the gain-of-function synucleinopathy hypothesis, which proposes that Lewy pathology causes Parkinson's disease: (1) most brains from people without neurological symptoms have multiple pathologies; (2) neither pathology type nor distribution correlate with disease severity or progression in Parkinson's disease; (3) aggregated α-synuclein in the form of Lewy bodies is not a space-occupying lesion but the insoluble fraction of its precursor, soluble monomeric α-synuclein; (4) pathology spread is passive, occurring by irreversible nucleation, not active replication; and (5) low cerebrospinal fluid α-synuclein levels predict brain atrophy and clinical disease progression. The transformation of α-synuclein into Lewy pathology may occur as a response to biological, toxic, or infectious stressors whose persistence perpetuates the nucleation process, depleting normal α-synuclein and eventually leading to Parkinson's symptoms from neuronal death. We propose testing the loss-of-function synucleinopenia hypothesis by evaluating the clinical and neurodegenerative rescue effect of replenishing the levels of monomeric α-synuclein.


Sujet(s)
Maladie de Parkinson , alpha-Synucléine , Animaux , Humains , alpha-Synucléine/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Corps de Lewy/anatomopathologie , Corps de Lewy/métabolisme , Maladie de Parkinson/métabolisme , Synucléinopathies/métabolisme , Synucléinopathies/anatomopathologie
19.
Ann Neurol ; 95(5): 843-848, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38501694

RÉSUMÉ

When effective treatments against neurodegenerative diseases become a reality, it will be important to know the age these pathologies begin to develop. We investigated alpha-synuclein pathology in brain tissue of the Tampere Sudden Death Study-unselected forensic autopsies on individuals living outside hospital institutions in Finland. Of 562 (16-95 years) participants, 42 were positive for Lewy-related pathology (LRP). The youngest LRP case was aged 54 years, and the frequency of LRP in individuals aged ≥50 years was 9%. This forensic autopsy study indicates LRP starts already in middle age and is more common than expected in the ≥50 years-of-age non-hospitalized population. ANN NEUROL 2024;95:843-848.


Sujet(s)
Mort subite , Maladie à corps de Lewy , alpha-Synucléine , Humains , Adulte d'âge moyen , Sujet âgé de 80 ans ou plus , Sujet âgé , Mâle , Femelle , Finlande/épidémiologie , Mort subite/anatomopathologie , Adolescent , Maladie à corps de Lewy/anatomopathologie , Maladie à corps de Lewy/métabolisme , alpha-Synucléine/métabolisme , Adulte , Jeune adulte , Encéphale/anatomopathologie , Encéphale/métabolisme , Autopsie , Corps de Lewy/anatomopathologie
20.
Alzheimers Dement ; 20(4): 2564-2574, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38353367

RÉSUMÉ

INTRODUCTION: Cerebral amyloid angiopathy (CAA) often accompanies dementia-associated pathologies and is important in the context of anti-amyloid monoclonal therapies and risk of hemorrhage. METHODS: We conducted a retrospective neuropathology-confirmed study of 2384 participants in the National Alzheimer Coordinating Center cohort (Alzheimer's disease [AD], n = 1175; Lewy body pathology [LBP], n = 316; and mixed AD and LBP [AD-LBP], n = 893). We used logistic regression to evaluate age, sex, education, APOE ε4, neuritic plaques, and neurofibrillary tangles (NFTs) in CAA risk. RESULTS: APOE ε4 increased CAA risk in all three groups, while younger age and higher NFT stages increased risk in AD and AD-LBP. In AD-LBP, male sex and lower education were additional risk factors. The odds of APOE ε4 carrier homozygosity related to CAA was higher in LBP (25.69) and AD-LBP (9.50) than AD (3.17). DISCUSSION: AD and LBPs modify risk factors for CAA and should be considered in reviewing the risk of CAA. HIGHLIGHTS: Lewy body pathology modifies risk factors for cerebral amyloid angiopathy (CAA) when present along with Alzheimer's disease (AD) neuropathology. In the context of anti-amyloid monoclonal therapies and their associated risks for hemorrhage, the risk of underlying CAA in mixed dementia with Lewy body pathology needs to be considered.


Sujet(s)
Maladie d'Alzheimer , Angiopathie amyloïde cérébrale , Mâle , Humains , Maladie d'Alzheimer/anatomopathologie , Apolipoprotéine E4/génétique , Corps de Lewy/anatomopathologie , Études rétrospectives , Angiopathie amyloïde cérébrale/épidémiologie , Angiopathie amyloïde cérébrale/anatomopathologie , Amyloïde , Facteurs de risque , Hémorragie , Plaque amyloïde/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE