Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 854
Filtrer
1.
Pharmacol Ther ; 260: 108683, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38950869

RÉSUMÉ

Parkinson's disease (PD) is diagnosed by its cardinal motor symptoms that are associated with the loss of dopamine neurons in the substantia nigra pars compacta (SNc). However, PD patients suffer from various non-motor symptoms years before diagnosis. These prodromal symptoms are thought to be associated with the appearance of Lewy body pathologies (LBP) in brainstem regions such as the dorsal motor nucleus of the vagus (DMV), the locus coeruleus (LC) and others. The neurons in these regions that are vulnerable to LBP are all slow autonomous pacemaker neurons that exhibit elevated oxidative stress due to their perpetual influx of Ca2+ ions. Aggregation of toxic α-Synuclein (aSyn) - the main constituent of LBP - during the long prodromal period challenges these vulnerable neurons, presumably altering their biophysics and physiology. In contrast to pathophysiology of late stage parkinsonism which is well-documented, little is known about the pathophysiology of the brainstem during prodromal PD. In this review, we discuss ion channel dysregulation associated with aSyn aggregation in brainstem pacemaker neurons and their cellular responses to them. While toxic aSyn elevates oxidative stress in SNc and LC pacemaker neurons and exacerbates their phenotype, DMV neurons mount an adaptive response that mitigates the oxidative stress. Ion channel dysregulation and cellular adaptations may be the drivers of the prodromal symptoms of PD. For example, selective targeting of toxic aSyn to DMV pacemakers, elevates the surface density of K+ channels, which slows their firing rate, resulting in reduced parasympathetic tone to the gastrointestinal tract, which resembles the prodromal PD symptoms of dysphagia and constipation. The divergent responses of SNc & LC vs. DMV pacemaker neurons may explain why the latter outlive the former despite presenting LBPs earlier. Elucidation the brainstem pathophysiology of prodromal PD could pave the way for physiological biomarkers, earlier diagnosis and novel neuroprotective therapies for PD.


Sujet(s)
Tronc cérébral , Canaux ioniques , Maladie de Parkinson , alpha-Synucléine , Humains , Animaux , Tronc cérébral/métabolisme , alpha-Synucléine/métabolisme , Maladie de Parkinson/métabolisme , Maladie de Parkinson/physiopathologie , Canaux ioniques/métabolisme , Stress oxydatif , Corps de Lewy/métabolisme
2.
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
3.
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
4.
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
5.
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 , alpha-Synucléine/métabolisme , alpha-Synucléine/génétique , Souris , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Corps de Lewy/métabolisme , Corps de Lewy/anatomopathologie , Vieillissement/génétique , Vieillissement/métabolisme , Modèles animaux de maladie humaine , Humains , Neurones dopaminergiques/métabolisme , Neurones dopaminergiques/anatomopathologie , Souris de lignée C57BL , Mâle
6.
Nat Commun ; 15(1): 3835, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38714700

RÉSUMÉ

Aggregated forms of α-synuclein constitute the major component of Lewy bodies, the proteinaceous aggregates characteristic of Parkinson's disease. Emerging evidence suggests that α-synuclein aggregation may occur within liquid condensates formed through phase separation. This mechanism of aggregation creates new challenges and opportunities for drug discovery for Parkinson's disease, which is otherwise still incurable. Here we show that the condensation-driven aggregation pathway of α-synuclein can be inhibited using small molecules. We report that the aminosterol claramine stabilizes α-synuclein condensates and inhibits α-synuclein aggregation within the condensates both in vitro and in a Caenorhabditis elegans model of Parkinson's disease. By using a chemical kinetics approach, we show that the mechanism of action of claramine is to inhibit primary nucleation within the condensates. These results illustrate a possible therapeutic route based on the inhibition of protein aggregation within condensates, a phenomenon likely to be relevant in other neurodegenerative disorders.


Sujet(s)
Caenorhabditis elegans , Maladie de Parkinson , Agrégats de protéines , alpha-Synucléine , alpha-Synucléine/métabolisme , alpha-Synucléine/composition chimique , Caenorhabditis elegans/métabolisme , Animaux , Maladie de Parkinson/métabolisme , Maladie de Parkinson/traitement médicamenteux , Humains , Agrégats de protéines/effets des médicaments et des substances chimiques , Agrégation pathologique de protéines/métabolisme , Agrégation pathologique de protéines/traitement médicamenteux , Modèles animaux de maladie humaine , Corps de Lewy/métabolisme , Cinétique
7.
J Biol Chem ; 300(6): 107315, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38663827

RÉSUMÉ

Lewy bodies (LB) are aberrant protein accumulations observed in the brain cells of individuals affected by Parkinson's disease (PD). A comprehensive analysis of LB proteome identified over a hundred proteins, many co-enriched with α-synuclein, a major constituent of LB. Within this context, OTUB1, a deubiquitinase detected in LB, exhibits amyloidogenic properties, yet the mechanisms underlying its aggregation remain elusive. In this study, we identify two critical sites in OTUB1-namely, positions 133 and 173-that significantly impact its amyloid aggregation. Substituting alanine at position 133 and lysine at position 173 enhances both thermodynamic and kinetic stability, effectively preventing amyloid aggregation. Remarkably, lysine at position 173 demonstrates the highest stability without compromising enzymatic activity. The increased stability and inhibition of amyloid aggregation are attributed mainly to the changes in the specific microenvironment at the hotspot. In our exploration of the in-vivo co-occurrence of α-synuclein and OTUB1 in LB, we observed a synergistic modulation of each other's aggregation. Collectively, our study unveils the molecular determinants influencing OTUB1 aggregation, shedding light on the role of specific residues in modulating aggregation kinetics and structural transition. These findings contribute valuable insights into the complex interplay of amino acid properties and protein aggregation, with potential implications for understanding broader aspects of protein folding and aggregation phenomena.


Sujet(s)
alpha-Synucléine , Humains , alpha-Synucléine/métabolisme , alpha-Synucléine/composition chimique , alpha-Synucléine/génétique , Cysteine endopeptidases/métabolisme , Cysteine endopeptidases/génétique , Cysteine endopeptidases/composition chimique , Enzymes de désubiquitinylation/métabolisme , Enzymes de désubiquitinylation/composition chimique , Agrégats de protéines , Corps de Lewy/métabolisme , Maladie de Parkinson/métabolisme , Maladie de Parkinson/génétique , Amyloïde/métabolisme , Amyloïde/composition chimique , Stabilité protéique , Stabilité enzymatique , Cinétique
8.
Nat Commun ; 15(1): 2436, 2024 Mar 18.
Article de Anglais | MEDLINE | ID: mdl-38499535

RÉSUMÉ

Parkinson's disease (PD) is closely linked to α-synuclein (α-syn) misfolding and accumulation in Lewy bodies. The PDZ serine protease HTRA1 degrades fibrillar tau, which is associated with Alzheimer's disease, and inactivating mutations to mitochondrial HTRA2 are implicated in PD. Here, we report that HTRA1 inhibits aggregation of α-syn as well as FUS and TDP-43, which are implicated in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. The protease domain of HTRA1 is necessary and sufficient for inhibiting aggregation, yet this activity is proteolytically-independent. Further, HTRA1 disaggregates preformed α-syn fibrils, rendering them incapable of seeding aggregation of endogenous α-syn, while reducing HTRA1 expression promotes α-syn seeding. HTRA1 remodels α-syn fibrils by targeting the NAC domain, the key domain catalyzing α-syn amyloidogenesis. Finally, HTRA1 detoxifies α-syn fibrils and prevents formation of hyperphosphorylated α-syn accumulations in primary neurons. Our findings suggest that HTRA1 may be a therapeutic target for a range of neurodegenerative disorders.


Sujet(s)
Maladie de Parkinson , alpha-Synucléine , Humains , alpha-Synucléine/génétique , alpha-Synucléine/métabolisme , Amyloïde/métabolisme , High-temperature requirement A serine peptidase 1/génétique , High-temperature requirement A serine peptidase 1/métabolisme , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Corps de Lewy/métabolisme
9.
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
10.
Cell Mol Life Sci ; 81(1): 75, 2024 Feb 05.
Article de Anglais | MEDLINE | ID: mdl-38315424

RÉSUMÉ

Autosomal dominant variants in LRP10 have been identified in patients with Lewy body diseases (LBDs), including Parkinson's disease (PD), Parkinson's disease-dementia (PDD), and dementia with Lewy bodies (DLB). Nevertheless, there is little mechanistic insight into the role of LRP10 in disease pathogenesis. In the brains of control individuals, LRP10 is typically expressed in non-neuronal cells like astrocytes and neurovasculature, but in idiopathic and genetic cases of PD, PDD, and DLB, it is also present in α-synuclein-positive neuronal Lewy bodies. These observations raise the questions of what leads to the accumulation of LRP10 in Lewy bodies and whether a possible interaction between LRP10 and α-synuclein plays a role in disease pathogenesis. Here, we demonstrate that wild-type LRP10 is secreted via extracellular vesicles (EVs) and can be internalised via clathrin-dependent endocytosis. Additionally, we show that LRP10 secretion is highly sensitive to autophagy inhibition, which induces the formation of atypical LRP10 vesicular structures in neurons in human-induced pluripotent stem cells (iPSC)-derived brain organoids. Furthermore, we show that LRP10 overexpression leads to a strong induction of monomeric α-synuclein secretion, together with time-dependent, stress-sensitive changes in intracellular α-synuclein levels. Interestingly, patient-derived astrocytes carrying the c.1424 + 5G > A LRP10 variant secrete aberrant high-molecular-weight species of LRP10 in EV-free media fractions. Finally, we show that this truncated patient-derived LRP10 protein species (LRP10splice) binds to wild-type LRP10, reduces LRP10 wild-type levels, and antagonises the effect of LRP10 on α-synuclein levels and distribution. Together, this work provides initial evidence for a possible functional role of LRP10 in LBDs by modulating intra- and extracellular α-synuclein levels, and pathogenic mechanisms linked to the disease-associated c.1424 + 5G > A LRP10 variant, pointing towards potentially important disease mechanisms in LBDs.


Sujet(s)
Maladie à corps de Lewy , Maladie de Parkinson , Humains , alpha-Synucléine/métabolisme , Maladie de Parkinson/anatomopathologie , Maladie à corps de Lewy/génétique , Maladie à corps de Lewy/métabolisme , Maladie à corps de Lewy/anatomopathologie , Corps de Lewy/métabolisme , Encéphale/métabolisme , Protéines apparentées au récepteur LDL/métabolisme
11.
J Parkinsons Dis ; 14(1): 17-33, 2024.
Article de Anglais | MEDLINE | ID: mdl-38189713

RÉSUMÉ

Lewy bodies (LBs) are pathological hallmarks of Parkinson's disease and dementia with Lewy bodies, characterized by the accumulation of α-synuclein (αSyn) protein in the brain. While LBs were first described a century ago, their formation and morphogenesis mechanisms remain incompletely understood. Here, we present a historical overview of LB definitions and highlight the importance of semantic clarity and precise definitions when describing brain inclusions. Recent breakthroughs in imaging revealed shared features within LB subsets and the enrichment of membrane-bound organelles in these structures, challenging the conventional LB formation model. We discuss the involvement of emerging concepts of liquid-liquid phase separation, where biomolecules demix from a solution to form dense condensates, as a potential LB formation mechanism. Finally, we emphasize the need for the operational definitions of LBs based on morphological characteristics and detection protocols, particularly in studies investigating LB formation mechanisms. A better understanding of LB organization and ultrastructure can contribute to the development of targeted therapeutic strategies for synucleinopathies.


Sujet(s)
Maladie à corps de Lewy , Maladie de Parkinson , Course à pied , Synucléinopathies , Humains , Corps de Lewy/métabolisme , Maladie de Parkinson/métabolisme , alpha-Synucléine/métabolisme , Encéphale/métabolisme , Synucléinopathies/métabolisme , Maladie à corps de Lewy/anatomopathologie
12.
J Theor Biol ; 581: 111734, 2024 03 21.
Article de Anglais | MEDLINE | ID: mdl-38246486

RÉSUMÉ

This paper presents a model for the growth of Lewy bodies (LBs), which are pathological hallmarks of Parkinson's disease (PD). The model simulates the growth of classical LBs, consisting of a core and a halo. The core is assumed to comprise lipid membrane fragments and damaged organelles, while the halo consists of radiating alpha-synuclein (α-syn) fibrils. The Finke-Watzky model is employed to simulate the aggregation of lipid fragments and α-syn monomers. Analytical and numerical exploration of the governing equations yielded approximate solutions applicable for larger times. The application of these approximate solutions to simulate LB radius growth led to the discovery of the cube root hypothesis, which posits that the LB radius is proportional to the cube root of its growth time. Sensitivity analysis revealed that the LB radius is unaffected by the kinetic rates of nucleation and autocatalytic growth, with growth primarily regulated by the production rates of lipid membrane fragments and α-syn monomers. The model indicates that the formation of large LBs associated with PD is dependent on the malfunction of the machinery responsible for the degradation of lipid membrane fragments, α-syn monomers, and their aggregates.


Sujet(s)
Corps de Lewy , Maladie de Parkinson , Humains , Corps de Lewy/métabolisme , Corps de Lewy/anatomopathologie , Radius/métabolisme , Radius/anatomopathologie , alpha-Synucléine/métabolisme , Maladie de Parkinson/métabolisme , Lipides
13.
Eur J Neurol ; 31(4): e16206, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38270442

RÉSUMÉ

BACKGROUND: Alpha-synuclein (α-Syn) oligomers and fibrils have been shown to augment the aggregation of TAR DNA-binding Protein 43 (TDP-43) monomers in vitro, supporting the idea that TDP-43 proteinopathies such as ALS may be modulated by the presence of toxic forms of α-Syn. Recently, parkinsonian features were reported in a study of European patients and Lewy bodies have been demonstrated pathologically in a similar series of patients. Based on these and other considerations, we sought to determine whether seed-competent α-Syn can be identified in spinal fluid of patients with ALS including familial, sporadic, and Guamanian forms of the disease. METHODS: Based on the finding that α-Syn has been found to be a prion-like protein, we have utilized a validated α-Synuclein seed amplification assay to determine if seed-competent α-Syn could be detected in the spinal fluid of patients with ALS. RESULTS: Toxic species of α-Syn were detected in CSF in 18 of 127 ALS patients, 5 of whom were from Guam. Two out of twenty six samples from patients with C9orf72 variant ALS had positive seed-amplification assays (SAAs). No positive tests were noted in superoxide dismutase type 1 ALS subjects (n = 14). The SAA was negative in 31 control subjects. CONCLUSIONS: Our findings suggest that a sub-group of ALS occurs in which self-replicating α-Syn is detectable and likely contributes to its pathogenesis. This finding may have implications for the diagnosis and treatment of this disorder.


Sujet(s)
Sclérose latérale amyotrophique , alpha-Synucléine , Humains , alpha-Synucléine/métabolisme , Sclérose latérale amyotrophique/anatomopathologie , Corps de Lewy/métabolisme , Corps de Lewy/anatomopathologie , Superoxide dismutase-1
14.
Nihon Yakurigaku Zasshi ; 159(1): 2-5, 2024.
Article de Japonais | MEDLINE | ID: mdl-38171833

RÉSUMÉ

The advent of a super-aged society poses urgent challenges in overcoming age-related neurological disorders and extending a healthy lifespan. Neurodegenerative diseases such as Alzheimer's disease, dementia with Lewy bodies, and Parkinson's disease are characterized by the accumulation of pathogenic proteins in the brain, leading to the formation of intracellular aggregates known as pathological hallmarks. In the early stages of protein accumulation, before the onset of clinical symptoms such as cognitive impairment or motor dysfunction, brain inflammation begins to occur. Subsequently, neuronal death progresses, and clinical symptoms manifest as dementia or Parkinson's disease. Therefore, there is a need for early prediction of neurodegeneration and the development of disease-modifying drugs for pre-symptomatic prevention. To address this issue, we have focused on enhancing the degradation of amyloid-ß protein by targeting Ca2+/calmodulin-dependent kinase II (CaMKII)/proteasome system and on suppressing the propagation and uptake mechanisms of α-synuclein by targeting fatty acid-binding proteins (FABPs) coupled with the long isoform of dopamine D2 (D2L) receptor. Additionally, our analysis of FABP knockout mice has revealed an increased expression of FABPs in the neurodegenerative process, suggesting their involvement in mitochondrial dysfunction and neuronal death. Based on these findings, this article highlights the physiological significance of FABP family proteins in neurodegeneration and discusses the analysis of plasma biomarkers for predicting neurodegenerative disorders and the discriminatory methods for distinguishing between Alzheimer's disease, dementia with Lewy bodies, and Parkinson's disease. Furthermore, we explore the potential of ultra-early prediction of neurodegenerative disorders.


Sujet(s)
Maladie d'Alzheimer , Maladie à corps de Lewy , Maladies neurodégénératives , Maladie de Parkinson , Animaux , Souris , Maladie à corps de Lewy/diagnostic , Maladie à corps de Lewy/anatomopathologie , Maladie de Parkinson/diagnostic , Maladie d'Alzheimer/métabolisme , Corps de Lewy/métabolisme , Corps de Lewy/anatomopathologie , alpha-Synucléine
15.
Int J Mol Sci ; 25(2)2024 Jan 21.
Article de Anglais | MEDLINE | ID: mdl-38279299

RÉSUMÉ

Parkinson's disease (PD) is a prevalent neurodegenerative disorder characterized by the progressive degeneration of dopaminergic neurons in the substantia nigra region of the brain. The hallmark pathological feature of PD is the accumulation of misfolded proteins, leading to the formation of intracellular aggregates known as Lewy bodies. Recent data evidenced how disruptions in protein synthesis, folding, and degradation are events commonly observed in PD and may provide information on the molecular background behind its etiopathogenesis. In the present study, we used a publicly available transcriptomic microarray dataset of peripheral blood of PD patients and healthy controls (GSE6613) to investigate the potential dysregulation of elements involved in proteostasis-related processes at the transcriptomic level. Our bioinformatics analysis revealed 375 differentially expressed genes (DEGs), of which 281 were down-regulated and 94 were up-regulated. Network analysis performed on the observed DEGs highlighted a cluster of 36 elements mainly involved in the protein synthesis processes. Different enriched ontologies were related to translation initiation and regulation, ribosome structure, and ribosome components nuclear export. Overall, this data consistently points to a generalized impairment of the translational machinery and proteostasis. Dysregulation of these mechanics has been associated with PD pathogenesis. Understanding the precise regulation of such processes may shed light on the molecular mechanisms of PD and provide potential data for early diagnosis.


Sujet(s)
Maladie de Parkinson , Humains , Maladie de Parkinson/métabolisme , Transcriptome , Corps de Lewy/métabolisme , Analyse de profil d'expression de gènes , Biosynthèse des protéines , Substantia nigra/métabolisme
16.
J Hazard Mater ; 465: 133312, 2024 03 05.
Article de Anglais | MEDLINE | ID: mdl-38147746

RÉSUMÉ

The emerging toxicant N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine quinone (6PPD-Q) is of wide concern due to its ubiquitous occurrence and high toxicity. Despite regular human exposure, limited evidence exists about its presence in the body and potential health risks. Herein, we analyzed cerebrospinal fluid (CSF) samples from Parkinson's disease (PD) patients and controls. The CSF levels of 6PPD-Q were twice as high in PD patients compared to controls. Immunostaining assays performed with primary dopaminergic neurons confirm that 6PPD-Q at environmentally relevant concentrations can exacerbate the formation of Lewy neurites induced by α-synuclein preformed fibrils (α-syn PFF). Assessment of cellular respiration reveals a considerable decrease in neuronal spare respiratory and ATP-linked respiration, potentially due to changes in mitochondrial membrane potential. Moreover, 6PPD-Q-induced mitochondrial impairment correlates with an upsurge in mitochondrial reactive oxygen species (mROS), and Mito-TEMPO-driven scavenging of mROS can lessen the amount of pathologic phospho-serine 129 α-synuclein. Untargeted metabolomics provides supporting evidence for the connection between 6PPD-Q exposure and changes in neuronal metabolite profiles. In-depth targeted metabolomics further unveils an overall reduction in glycolysis metabolite pool and fluctuations in the quantity of TCA cycle intermediates. Given its potentially harmful attributes, the presence of 6PPD-Q in human brain could potentially be a risk factor for PD.


Sujet(s)
Maladies mitochondriales , Maladie de Parkinson , Humains , alpha-Synucléine/métabolisme , Neurones dopaminergiques , Corps de Lewy/métabolisme , Corps de Lewy/anatomopathologie , Maladies mitochondriales/métabolisme , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Quinones/métabolisme
17.
Mol Neurodegener ; 18(1): 91, 2023 Nov 27.
Article de Anglais | MEDLINE | ID: mdl-38012703

RÉSUMÉ

BACKGROUND: Alpha-synuclein (α-syn) aggregation into proteinaceous intraneuronal inclusions, called Lewy bodies (LBs), is the neuropathological hallmark of Parkinson's disease (PD) and related synucleinopathies. However, the exact role of α-syn inclusions in PD pathogenesis remains elusive. This lack of knowledge is mainly due to the absence of optimal α-syn-based animal models that recapitulate the different stages of neurodegeneration. METHODS: Here we describe a novel approach for a systemic delivery of viral particles carrying human α-syn allowing for a large-scale overexpression of this protein in the mouse brain. This approach is based on the use of a new generation of adeno-associated virus (AAV), AAV-PHP.eB, with an increased capacity to cross the blood-brain barrier, thus offering a viable tool for a non-invasive and large-scale gene delivery in the central nervous system. RESULTS: Using this model, we report that widespread overexpression of human α-syn induced selective degeneration of dopaminergic (DA) neurons, an exacerbated neuroinflammatory response in the substantia nigra and a progressive manifestation of PD-like motor impairments. Interestingly, biochemical analysis revealed the presence of insoluble α-syn oligomers in the midbrain. Together, our data demonstrate that a single non-invasive systemic delivery of viral particles overexpressing α-syn prompted selective and progressive neuropathology resembling the early stages of PD. CONCLUSIONS: Our new in vivo model represents a valuable tool to study the role of α-syn in PD pathogenesis and in the selective vulnerability of nigral DA neurons; and offers the opportunity to test new strategies targeting α-syn toxicity for the development of disease-modifying therapies for PD and related disorders.


Sujet(s)
Maladie de Parkinson , Souris , Animaux , Humains , Maladie de Parkinson/métabolisme , alpha-Synucléine/métabolisme , Rodentia/métabolisme , Encéphale/métabolisme , Corps de Lewy/métabolisme , Substantia nigra/anatomopathologie , Neurones dopaminergiques/métabolisme , Modèles animaux de maladie humaine
18.
Sci Rep ; 13(1): 19020, 2023 11 03.
Article de Anglais | MEDLINE | ID: mdl-37923923

RÉSUMÉ

Protein misfolding and aggregation play crucial roles in amyloidogenic diseases through the self-assembly of intrinsically disordered proteins (IDPs) in type II diabetes (T2D), Alzheimer's disease (AD) and Parkinson's disease (PD). PD is the most common neurodegenerative disorder after AD, and is associated with the loss of dopaminergic signaling, which causes motor and nonmotor signs and symptoms. Lewy bodies and Lewy neurites are common pathological hallmarks of PD that are mainly composed of aggregates of disordered α-synuclein (α-Syn). There have been many efforts to develop chemical compounds to prevent aggregation or facilitate disruption of the aggregates. Furthermore, the roles and interactions of many compounds have yet to be revealed at the atomistic level, especially their impacts on the dynamics and chain-chain interactions of the oligomers, which are of interest in this study. The conformational diversity and detailed interactions among homo-oligomer chains of α-Syn are not fully discovered; identifying these might help uncover a practical approach to developing a potent therapy. In this study, we used an in-silico investigation to address the conformational diversity of α-Syn oligomer. The roles of several point mutations in protein aggregation in PD are known; we take this further by evaluating the interaction energies and contributions of all residues in stability and residue-chain interactions. In this study, we docked chemical derivatives of three compounds with high drug-likeness properties to evaluate the roles of our ligands in the conformational dynamicity of the oligomers, with emphasis on intramolecular forces. Free energy evaluation of the modeled inter and intramolecular interactions through MD simulation shows effective interaction and binding between α-Syn and our compounds. However, we find that they do not significantly disrupt the chain-chain interactions, compared to unliganded simulation.


Sujet(s)
Maladie d'Alzheimer , Diabète de type 2 , Maladie de Parkinson , Humains , alpha-Synucléine/métabolisme , Diabète de type 2/métabolisme , Maladie de Parkinson/métabolisme , Corps de Lewy/métabolisme , Maladie d'Alzheimer/métabolisme
19.
Nat Commun ; 14(1): 6892, 2023 10 28.
Article de Anglais | MEDLINE | ID: mdl-37898614

RÉSUMÉ

Extraction of α-Synuclein (αSyn) aggregates from Lewy body disease (LBD) brains has been widely described yet templated fibrillization of LB-αSyn often fails to propagate its structural and functional properties. We recently demonstrated that aggregates amplified from LB-αSyn (ampLB) show distinct biological activities in vitro compared to human αSyn preformed fibrils (hPFF) formed de novo. Here we compare the in vivo biological activities of hPFF and ampLB regarding seeding activity, latency in inducing pathology, distribution of pathology, inclusion morphology, and cell-type preference. Injection of ampLB into mice expressing only human αSyn (male Thy1:SNCA/Snca-/- mice) induced pathologies similar to those of LBD subjects that were distinct from those induced by hPFF-injection or developing spontaneously with aging. Importantly, αSyn aggregates in ampLB-injected Thy1:SNCA/Snca-/- mice maintained the unique biological and conformational features of original LB-αSyn. These results indicate that ampLB-injection, rather than conventional PFF-injection or αSyn overexpression, faithfully models key aspects of LBD.


Sujet(s)
Maladie à corps de Lewy , Souris , Mâle , Humains , Animaux , Maladie à corps de Lewy/anatomopathologie , alpha-Synucléine/métabolisme , Corps de Lewy/métabolisme , Encéphale/métabolisme , Vieillissement
20.
Mol Brain ; 16(1): 72, 2023 10 17.
Article de Anglais | MEDLINE | ID: mdl-37848910

RÉSUMÉ

The major neuropathologic feature of Parkinson's disease is the presence of widespread intracellular inclusions of α-synuclein known as Lewy bodies. Evidence suggests that these misfolded protein inclusions spread through the brain with disease progression. Changes in synaptic function precede neurodegeneration, and this extracellular α-synuclein can affect synaptic transmission. However, whether and how the spreading of α-synuclein aggregates modulates synaptic function before neuronal loss remains unknown. In the present study, we investigated the effect of intrastriatal injection of α-synuclein preformed fibrils (PFFs) on synaptic activity in the somatosensory cortex using a combination of whole-cell patch-clamp electrophysiology, histology, and Golgi-Cox staining. Intrastriatal PFF injection was followed by formation of phosphorylated α-synuclein inclusions in layer 5 of the somatosensory cortex, leading to a decrease in synapse density, dendritic spines, and spontaneous excitatory post-synaptic currents, without apparent neuronal loss. Additionally, three-dimensional reconstruction of microglia using confocal imaging showed an increase in the engulfment of synapses. Collectively, our data indicate that propagation of α-synuclein through neural networks causes abnormalities in synaptic structure and dynamics prior to neuronal loss.


Sujet(s)
Maladie de Parkinson , alpha-Synucléine , Humains , alpha-Synucléine/métabolisme , Microglie/métabolisme , Maladie de Parkinson/anatomopathologie , Corps de Lewy/métabolisme , Synapses/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE