Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 124
Filtrer
1.
Nat Commun ; 15(1): 7619, 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39223119

RÉSUMÉ

Isolated REM Sleep Behavior Disorder (iRBD) is considered a prodrome of Parkinson's disease (PD). We investigate whether the potentially disease-modifying compound acetyl-DL-leucine (ADLL; 5 g/d) has an effect on prodromal PD progression in 2 iRBD-patients. Outcome parameters are RBD-severity sum-score (RBD-SS-3), dopamine-transporter single-photon emission computerized tomography (DAT-SPECT) and metabolic "Parkinson-Disease-related-Pattern (PDRP)"-z-score in 18F-fluorodeoxyglucose positron emission tomography (FDG-PET). After 3 weeks ADLL-treatment, the RBD-SS-3 drops markedly in both patients and remains reduced for >18 months of ADLL-treatment. In patient 1 (female), the DAT-SPECT putaminal binding ratio (PBR) decreases in the 5 years pretreatment from normal (1.88) to pathological (1.22) and the patient's FDG-PET-PDRP-z-score rises from 1.72 to 3.28 (pathological). After 22 months of ADLL-treatment, the DAT-SPECT-PBR increases to 1.67 and the FDG-PET-PDRP-z-score stabilizes at 3.18. Similar results are seen in patient 2 (male): his DAT-SPECT-PBR rises from a pretreatment value of 1.42 to 1.72 (close to normal) and the FDG-PET-PDRP-z-score decreases from 1.02 to 0.30 after 18 months of ADLL-treatment. These results support exploration of whether ADLL may have disease-modifying properties in prodromal PD.


Sujet(s)
Transporteurs de la dopamine , Leucine , Maladie de Parkinson , Tomographie par émission de positons , Trouble du comportement en sommeil paradoxal , Tomographie par émission monophotonique , Humains , Femelle , Trouble du comportement en sommeil paradoxal/métabolisme , Trouble du comportement en sommeil paradoxal/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/traitement médicamenteux , Mâle , Transporteurs de la dopamine/métabolisme , Maladie de Parkinson/métabolisme , Maladie de Parkinson/imagerie diagnostique , Maladie de Parkinson/traitement médicamenteux , Maladie de Parkinson/anatomopathologie , Tomographie par émission de positons/méthodes , Leucine/métabolisme , Leucine/analogues et dérivés , Sujet âgé , Adulte d'âge moyen , Encéphale/métabolisme , Encéphale/imagerie diagnostique , Encéphale/anatomopathologie , Fluorodésoxyglucose F18/métabolisme , Corps strié/métabolisme , Corps strié/imagerie diagnostique
2.
J Parkinsons Dis ; 14(s2): S323-S331, 2024.
Article de Anglais | MEDLINE | ID: mdl-38995801

RÉSUMÉ

Synucleinopathies are disorders characterized by the aggregation and deposition of pathological α-synuclein conformers. The underlying neurodegenerative processes begin years or decades before the onset of cardinal motor symptoms. This prodromal phase may manifest with various signs or symptoms. However, there are no current standardized laboratory tests to ascertain the progression and conversion of prodromal conditions such as mild cognitive impairment, isolated REM sleep behavior disorder or pure autonomic failure. The aim of this systematic review was to evaluate the diagnostic possibilities using human biofluids as source material to detect pathological α-synuclein in the prodromal phase of synucleinopathies. Our review identified eight eligible studies, that investigated pathological α-synuclein conformers using cerebrospinal fluid from patients with prodromal signs of synulceinopathies to differentiate this patient group from non-synucleinopathies, while only one study investigated this aspect using blood as medium. While previous studies clearly demonstrated a high diagnostic performance of α-synuclein seed amplification assays for differentiating synucleinopathies with Lewy bodies from healthy controls, only few analyses were performed focussing on individuals with prodromal disease. Nevertheless, results for the early detection of α-synuclein seeds using α-synuclein seed amplification assays were promising and may be of particular relevance for future clinical trials and clinical practice.


Sujet(s)
Symptômes prodromiques , Synucléinopathies , alpha-Synucléine , Humains , alpha-Synucléine/métabolisme , alpha-Synucléine/liquide cérébrospinal , Synucléinopathies/diagnostic , Synucléinopathies/métabolisme , Marqueurs biologiques/liquide cérébrospinal , Marqueurs biologiques/métabolisme , Trouble du comportement en sommeil paradoxal/diagnostic , Trouble du comportement en sommeil paradoxal/métabolisme , Dysfonctionnement cognitif/diagnostic , Dysfonctionnement cognitif/métabolisme
3.
Mov Disord ; 39(8): 1300-1309, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38715177

RÉSUMÉ

OBJECTIVE: Evidence of abnormal α-synuclein (α-Syn) deposition in the brain is required for definitive diagnosis of synucleinopathies, which remains challenging. The seed amplification assay (SAA) is an innovative technique that can detect the seeding activity of misfolded α-Syn, enabling the amplification and detection of minute quantities of pathogenic α-Syn aggregates. This study aimed to evaluate oral mucosa α-Syn SAA as possible diagnostic and prodromal biomarkers for synucleinopathies. METHODS: A total of 107 Parkinson's disease (PD) patients, 99 multiple system atrophy (MSA) patients, 33 patients with isolated rapid eye movement sleep behavior disorder (iRBD) and 103 healthy controls (HC) were included. The SAA was applied to detect the seeding activity of α-Syn from oral mucosa. A combination of morphological, biochemical, and biophysical methods was also used to analyze the fibrils generated from the oral mucosa α-Syn SAA. RESULTS: Structured illumination microscopy images revealed the increased α-Syn species in oral mucosa of PD, MSA, and iRBD patients than in HCs. Oral mucosa α-Syn SAA distinguished patients with PD from HC with 67.3% sensitivity and 90.3% specificity. Oral mucosa was α-Syn SAA positive in 53.5% MSA patients and 63.6% iRBD patients. Furthermore, the α-Syn fibrils generated from MSA demonstrated greater resistance to proteinase K digestion and exhibited stronger cytotoxicity compared to those from PD patients. CONCLUSION: Oral mucosa α-Syn seeding activity may serve as novel non-invasive diagnostic and prodromal biomarkers for synucleinopathies. The α-Syn aggregates amplified from the oral mucosa of PD and MSA exhibited distinct biochemical and biophysical properties. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Sujet(s)
Muqueuse de la bouche , Atrophie multisystématisée , Maladie de Parkinson , Trouble du comportement en sommeil paradoxal , Synucléinopathies , alpha-Synucléine , Humains , Trouble du comportement en sommeil paradoxal/métabolisme , Trouble du comportement en sommeil paradoxal/diagnostic , alpha-Synucléine/métabolisme , Femelle , Mâle , Maladie de Parkinson/métabolisme , Maladie de Parkinson/diagnostic , Adulte d'âge moyen , Sujet âgé , Synucléinopathies/métabolisme , Muqueuse de la bouche/métabolisme , Muqueuse de la bouche/anatomopathologie , Atrophie multisystématisée/métabolisme , Atrophie multisystématisée/diagnostic , Atrophie multisystématisée/génétique , Atrophie multisystématisée/anatomopathologie , Marqueurs biologiques/métabolisme
4.
Mov Disord ; 39(8): 1397-1402, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38561921

RÉSUMÉ

BACKGROUND: Idiopathic rapid eye movement sleep behavior disorder (iRBD) is considered as a prodromal stage of synucleinopathies. Fecal short-chain fatty acid (SCFA) changes in iRBD and the relationships with synucleinopathies have never been investigated. OBJECTIVES: To investigate fecal SCFA changes among iRBD, multiple system atrophy (MSA), and Parkinson's disease (PD), and evaluate their relationships. METHODS: Fecal SCFAs and gut microbiota were measured in 29 iRBD, 42 MSA, 40 PD, and 35 normal controls (NC) using gas chromatography-mass spectrometry and 16S rRNA gene sequencing. RESULTS: Compared with NC, fecal SCFA levels (propionic, acetic, and butyric acid) were lower in iRBD, MSA, and PD. Combinations of these SCFAs could differentiate NC from iRBD (AUC 0.809), MSA (AUC 0.794), and PD (AUC 0.701). Decreased fecal SCFAs were associated with the common reducing SCFA-producing gut microbiota in iRBD, MSA, and PD. CONCLUSIONS: iRBD shares similar fecal SCFA alterations with MSA and PD, and the combination of these SCFAs might be a potential synucleinopathies-related biomarker. © 2024 International Parkinson and Movement Disorder Society.


Sujet(s)
Acides gras volatils , Fèces , Microbiome gastro-intestinal , Atrophie multisystématisée , Maladie de Parkinson , Trouble du comportement en sommeil paradoxal , Humains , Atrophie multisystématisée/métabolisme , Trouble du comportement en sommeil paradoxal/métabolisme , Maladie de Parkinson/métabolisme , Fèces/composition chimique , Fèces/microbiologie , Mâle , Femelle , Acides gras volatils/métabolisme , Acides gras volatils/analyse , Sujet âgé , Adulte d'âge moyen , Microbiome gastro-intestinal/physiologie
5.
J Parkinsons Dis ; 14(4): 823-831, 2024.
Article de Anglais | MEDLINE | ID: mdl-38640171

RÉSUMÉ

Background: Rapid eye movement sleep behavior disorder (RBD) may precede or follow motor symptoms in Parkinson's disease (PD). While over 70% of idiopathic RBD cases phenoconvert within a decade, a small subset develops PD after a more extended period or remains nonconverted. These heterogeneous manifestations of RBD in PD prompt subtype investigations. Premotor RBD may signify "body-first" PD with bottom-up, symmetric synucleinopathy propagation. Objective: Explore brainstem and nigrostriatal monoaminergic degeneration pattern differences based on premotor RBD presence and duration in de novo PD patients. Methods: In a cross-sectional analysis of de novo PD patients (n = 150) undergoing FP-CIT PET and RBD Single-Question Screen, the cohort was categorized into groups with and without premotor RBD (PDRBD +/-), with further classification of PDRBD + based on a 10-year duration of premotor RBD. Analysis of FP-CIT binding in the striatum and pons, striatal asymmetry, and striatum-to-pons ratios compared patterns of nigrostriatal and brainstem monoaminergic degeneration. Results: PDRBD + exhibited more severe and symmetrical striatal dopaminergic denervation compared to PDRBD-, with the difference in severity accentuated in the least-affected hemisphere. The PDRBD +<10Y subgroup displayed the most prominent striatal symmetry, supporting a more homogeneous "body-first" subtype. Pontine uptakes remained lower in PDRBD + even after adjusting for striatal uptake, suggesting early degeneration of pontine monoaminergic nuclei. Conclusions: Premotor RBD in PD is associated with severe, symmetrical nigrostriatal and brainstem monoaminergic degeneration, especially in cases with PD onset within 10 years of RBD. This supports the concept of a "widespread, bottom-up" pathophysiological mechanism associated with premotor RBD in PD.


Sujet(s)
Maladie de Parkinson , Tomographie par émission de positons , Trouble du comportement en sommeil paradoxal , Humains , Trouble du comportement en sommeil paradoxal/métabolisme , Trouble du comportement en sommeil paradoxal/étiologie , Trouble du comportement en sommeil paradoxal/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/anatomopathologie , Maladie de Parkinson/complications , Maladie de Parkinson/métabolisme , Maladie de Parkinson/imagerie diagnostique , Maladie de Parkinson/anatomopathologie , Maladie de Parkinson/physiopathologie , Mâle , Sujet âgé , Femelle , Adulte d'âge moyen , Études transversales , Corps strié/métabolisme , Corps strié/imagerie diagnostique , Corps strié/anatomopathologie , Tronc cérébral/imagerie diagnostique , Tronc cérébral/métabolisme , Tronc cérébral/anatomopathologie , Tropanes , Substantia nigra/imagerie diagnostique , Substantia nigra/métabolisme , Substantia nigra/anatomopathologie
6.
Sleep Med ; 119: 135-138, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38678756

RÉSUMÉ

OBJECTIVE/BACKGROUND: Idiopathic/isolated REM sleep behavior disorder (iRBD) is widely regarded as an early sign of neurodegeneration leading to synucleinopathies. While circadian rhythm alterations in iRBD have been preliminarily demonstrated, evidence on melatonin secretion patterns in this clinical condition is limited. To address this knowledge gap, this exploratory study aimed to integrate salivary melatonin measurement with actigraphic monitoring in individuals with iRBD and age-matched healthy controls (HC) under real-life conditions. METHODS: Participants diagnosed with iRBD and HC underwent clinical evaluation and wore an actigraph for seven days and nights. Salivary melatonin concentrations were measured at five time points during the last night of recording. Comparative analyses were conducted on clinical data, actigraphic parameters, and melatonin levels between the two groups. RESULTS: iRBD participants (n = 18) showed greater motor (p < 0.01) and non-motor symptoms (p < 0.001), alongside disruptions in circadian sleep-wake rhythm compared to HC (n = 10). Specifically, actigraphy revealed a delayed central phase measurement (p < 0.05), reduced activity during the most active hours (p < 0.001), and decreased relative amplitude (p < 0.05). Total salivary melatonin concentration was significantly lower in iRBD (p < 0.05), with a slight but non-significant phase delay in dim light melatonin onset. CONCLUSIONS: This exploratory study highlights a dysregulation of circadian sleep-wake rhythm coupled with reduced melatonin secretion in iRBD. Future research could add to these preliminary findings to evaluate novel treatment approaches to regulate the sleep-wake cycle and elucidate the implications of circadian dysregulation in the conversion from iRBD to neurodegeneration.


Sujet(s)
Actigraphie , Rythme circadien , Mélatonine , Trouble du comportement en sommeil paradoxal , Salive , Humains , Mélatonine/métabolisme , Mélatonine/analyse , Salive/composition chimique , Salive/métabolisme , Mâle , Trouble du comportement en sommeil paradoxal/métabolisme , Trouble du comportement en sommeil paradoxal/physiopathologie , Femelle , Rythme circadien/physiologie , Sujet âgé , Adulte d'âge moyen
7.
Eur J Nucl Med Mol Imaging ; 50(11): 3290-3301, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37310428

RÉSUMÉ

PURPOSE: Isolated REM sleep behaviour disorder (iRBD) patients are at high risk of developing clinical syndromes of the α-synuclein spectrum. Progression markers are needed to determine the neurodegenerative changes and to predict their conversion. Brain imaging with 18F-FDG PET in iRBD is promising, but longitudinal studies are scarce. We investigated the regional brain changes in iRBD over time, related to phenoconversion. METHODS: Twenty iRBD patients underwent two consecutive 18F-FDG PET brain scans and clinical assessments (3.7 ± 0.6 years apart). Seventeen patients also underwent 123I-MIBG and 123I-FP-CIT SPECT scans at baseline. Four subjects phenoconverted to Parkinson's disease (PD) during follow-up. 18F-FDG PET scans were compared to controls with a voxel-wise single-subject procedure. The relationship between regional brain changes in metabolism and PD-related pattern scores (PDRP) was investigated. RESULTS: Individual hypometabolism t-maps revealed three scenarios: (1) normal 18F-FDG PET scans at baseline and follow-up (N = 10); (2) normal scans at baseline but occipital or occipito-parietal hypometabolism at follow-up (N = 4); (3) occipital hypometabolism at baseline and follow-up (N = 6). All patients in the last group had pathological 123I-MIBG and 123I-FP-CIT SPECT. iRBD converters (N = 4) showed occipital hypometabolism at baseline (third scenario). At the group level, hypometabolism in the frontal and occipito-parietal regions and hypermetabolism in the cerebellum and limbic regions were progressive over time. PDRP z-scores increased over time (0.54 ± 0.36 per year). PDRP expression was driven by occipital hypometabolism and cerebellar hypermetabolism. CONCLUSIONS: Our results suggest that occipital hypometabolism at baseline in iRBD implies a short-term conversion to PD. This might help in stratification strategies for disease-modifying trials.


Sujet(s)
Maladie de Parkinson , Trouble du comportement en sommeil paradoxal , Humains , Trouble du comportement en sommeil paradoxal/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/métabolisme , Maladie de Parkinson/imagerie diagnostique , Maladie de Parkinson/métabolisme , Fluorodésoxyglucose F18 , 3-Iodobenzyl-guanidine , Tomographie par émission de positons/méthodes , Facteurs de risque
8.
Transl Neurodegener ; 12(1): 27, 2023 05 22.
Article de Anglais | MEDLINE | ID: mdl-37217951

RÉSUMÉ

BACKGROUND: The isolated rapid-eye-movement sleep behavior disorder (iRBD) is a prodromal condition of Lewy body disease including Parkinson's disease and dementia with Lewy bodies (DLB). We aim to investigate the longitudinal evolution of DLB-related cortical thickness signature in a prospective iRBD cohort and evaluate the possible predictive value of the cortical signature index in predicting dementia-first phenoconversion in individuals with iRBD. METHODS: We enrolled 22 DLB patients, 44 healthy controls, and 50 video polysomnography-proven iRBD patients. Participants underwent 3-T magnetic resonance imaging (MRI) and clinical/neuropsychological evaluations. We characterized DLB-related whole-brain cortical thickness spatial covariance pattern (DLB-pattern) using scaled subprofile model of principal components analysis that best differentiated DLB patients from age-matched controls. We analyzed clinical and neuropsychological correlates of the DLB-pattern expression scores and the mean values of the whole-brain cortical thickness in DLB and iRBD patients. With repeated MRI data during the follow-up in our prospective iRBD cohort, we investigated the longitudinal evolution of the cortical thickness signature toward Lewy body dementia. Finally, we analyzed the potential predictive value of cortical thickness signature as a biomarker of phenoconversion in iRBD cohort. RESULTS: The DLB-pattern was characterized by thinning of the temporal, orbitofrontal, and insular cortices and relative preservation of the precentral and inferior parietal cortices. The DLB-pattern expression scores correlated with attentional and frontal executive dysfunction (Trail Making Test-A and B: R = - 0.55, P = 0.024 and R = - 0.56, P = 0.036, respectively) as well as visuospatial impairment (Rey-figure copy test: R = - 0.54, P = 0.0047). The longitudinal trajectory of DLB-pattern revealed an increasing pattern above the cut-off in the dementia-first phenoconverters (Pearson's correlation, R = 0.74, P = 6.8 × 10-4) but no significant change in parkinsonism-first phenoconverters (R = 0.0063, P = 0.98). The mean value of the whole-brain cortical thickness predicted phenoconversion in iRBD patients with hazard ratio of 9.33 [1.16-74.12]. The increase in DLB-pattern expression score discriminated dementia-first from parkinsonism-first phenoconversions with 88.2% accuracy. CONCLUSION: Cortical thickness signature can effectively reflect the longitudinal evolution of Lewy body dementia in the iRBD population. Replication studies would further validate the utility of this imaging marker in iRBD.


Sujet(s)
Maladie à corps de Lewy , Maladie de Parkinson , Syndromes parkinsoniens , Trouble du comportement en sommeil paradoxal , Humains , Trouble du comportement en sommeil paradoxal/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/épidémiologie , Trouble du comportement en sommeil paradoxal/métabolisme , Maladie à corps de Lewy/imagerie diagnostique , Études prospectives
9.
Int J Mol Sci ; 24(10)2023 May 16.
Article de Anglais | MEDLINE | ID: mdl-37240185

RÉSUMÉ

REM sleep behavior disorder (RBD) has a tighter link with synucleinopathies than other neurodegenerative disorders. Parkinson's Disease (PD) patients with RBD have a more severe motor and cognitive impairment; biomarkers for RBD are currently unavailable. Synaptic accumulation of α-Syn oligomers and their interaction with SNARE proteins is responsible for synaptic dysfunction in PD. We verified whether oligomeric α-Syn and SNARE components in neural-derived extracellular vesicles (NDEVs) in serum could be biomarkers for RBD. Forty-seven PD patients were enrolled, and the RBD Screening Questionnaire (RBDSQ) was compiled. A cut-off score > 6 to define probable RBD (p-RBD) and probable non-RBD (p non-RBD) was used. NDEVs were isolated from serum by immunocapture, and oligomeric α-Syn and SNARE complex components VAMP-2 and STX-1 were measured by ELISA. NDEVs' STX-1A resulted in being decreased in p-RBD compared to p non-RBD PD patients. A positive correlation between NDEVs' oligomeric α-Syn and RBDSQ total score was found (p = 0.032). Regression analysis confirmed a significant association between NDEVs' oligomeric α-Syn concentration and RBD symptoms (p = 0.033) independent from age, disease duration, and motor impairment severity. Our findings suggest that synuclein-mediated neurodegeneration in PD-RBD is more diffuse. NDEVs' oligomeric α-Syn and SNARE complex components' serum concentrations could be regarded as reliable biomarkers for the RBD-specific PD endophenotype.


Sujet(s)
Maladie de Parkinson , Trouble du comportement en sommeil paradoxal , Humains , Maladie de Parkinson/métabolisme , alpha-Synucléine/métabolisme , Trouble du comportement en sommeil paradoxal/diagnostic , Trouble du comportement en sommeil paradoxal/étiologie , Trouble du comportement en sommeil paradoxal/métabolisme , Études de cohortes , Enquêtes et questionnaires , Marqueurs biologiques
10.
J Neurol Neurosurg Psychiatry ; 94(7): 532-540, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-36725328

RÉSUMÉ

BACKGROUND: Isolated rapid eye movement (REM) sleep behaviour disorder (iRBD) is a prodromal state of clinical α-synucleinopathies such as Parkinson's disease and Lewy body dementia. The lead-time until conversion is unknown. The most reliable marker of progression is reduced striatal dopamine transporter (DAT) binding, but low availability of imaging facilities limits general use. Our prospective observational study aimed to relate metrics of REM sleep without atonia (RWA)-a hallmark of RBD-to DAT-binding ratios in a large, homogeneous sample of patients with RBD to explore the utility of RWA as a marker of progression in prodromal α-synucleinopathies. METHODS: DAT single-photon emission CT (SPECT) and video polysomnography (vPSG) were performed in 221 consecutive patients with clinically suspected RBD. RESULTS: vPSG confirmed RBD in 176 patients (162 iRBD, 14 phenoconverted, 45 non-synucleinopathies). Specific DAT-binding ratios differed significantly between groups, but showed considerable overlap. Most RWA metrics correlated significantly with DAT-SPECT ratios (eg, Montreal tonic vs most-affected-region: r=-0.525; p<0.001). In patients taking serotonergic/noradrenergic antidepressants or dopaminergic substances or with recent alcohol abuse, correlations were weaker, suggesting a confounding influence, unlike other possible confounders such as beta-blocker use or comorbid sleep apnoea. CONCLUSIONS: In this large single-centre prospective observational study, we found evidence that DAT-binding ratios in patients with iRBD can be used to describe a continuum in the neurodegenerative process. Overlap with non-synucleinopathies and clinical α-synucleinopathies, however, precludes the use of DAT-binding ratios as a precise diagnostic marker. The parallel course of RWA metrics and DAT-binding ratios suggests in addition to existing data that RWA, part of the routine diagnostic workup in these patients, may represent a marker of progression. Based on our findings, we suggest ranges of RWA values to estimate whether patients are in an early, medium or advanced state within the prodromal phase of α-synucleinopathies, providing them with important information about time until possible conversion.


Sujet(s)
Maladie à corps de Lewy , Trouble du comportement en sommeil paradoxal , Synucléinopathies , Humains , Synucléinopathies/diagnostic , Sommeil paradoxal , Pronostic , Maladie à corps de Lewy/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/métabolisme , Protéines de transport membranaire , Marqueurs biologiques
11.
Mov Disord ; 38(4): 567-578, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36781413

RÉSUMÉ

BACKGROUND: Misfolded α-synuclein (αSyn) aggregates (αSyn-seeds) in cerebrospinal fluid (CSF) are biomarkers for synucleinopathies such as Parkinson's disease (PD). αSyn-seeds have been detected in prodromal cases with isolated rapid eye movement sleep behavior disorder (iRBD). OBJECTIVES: The objective of this study was to determine the accuracy of the αSyn-seed amplification assay (αS-SAA) in a comprehensively characterized cohort with a high proportion of PD and iRBD CSF samples collected at baseline. METHODS: We used a high-throughput αS-SAA to analyze 233 blinded CSF samples from 206 participants of the DeNovo Parkinson Cohort (DeNoPa) (113 de novo PD, 64 healthy controls, 29 iRBD confirmed by video polysomnography). Results were compared with the final diagnosis, which was determined after up to 10 years of longitudinal clinical evaluations, including dopamine-transporter-single-photon emission computed tomography (DAT-SPECT) at baseline, CSF proteins, Movement Disorder Society-Unified Parkinson's Disease Rating Scale, and various cognitive and nonmotor scales. RESULTS: αS-SAA detected αSyn-seeds in baseline PD-CSF with 98% accuracy. αSyn-seeds were detected in 93% of the iRBD cases. αS-SAA results showed higher agreement with the final than the initial diagnosis, as 14 patients were rediagnosed as non-αSyn aggregation disorder. For synucleinopathies, αS-SAA showed higher concordance with the final diagnosis than DAT-SPECT. Statistically significant correlations were found between assay parameters and disease progression. CONCLUSIONS: Our results confirm αS-SAA accuracy at the first clinical evaluation when a definite diagnosis is most consequential. αS-SAA conditions reported here are highly sensitive, enabling the detection of αSyn-seeds in CSF from iRBD just months after the first symptoms, suggesting that αSyn-seeds are present in the very early prodromal phase of synucleinopathies. Therefore, αSyn-seeds are clear risk markers for synuclein-related disorders, but not for time of phenoconversion. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Sujet(s)
Maladie de Parkinson , Trouble du comportement en sommeil paradoxal , alpha-Synucléine , Humains , alpha-Synucléine/liquide cérébrospinal , alpha-Synucléine/composition chimique , Maladie de Parkinson/diagnostic , Maladie de Parkinson/métabolisme , Trouble du comportement en sommeil paradoxal/diagnostic , Trouble du comportement en sommeil paradoxal/métabolisme , Synucléinopathies/diagnostic
12.
Mov Disord ; 38(1): 57-67, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36190111

RÉSUMÉ

BACKGROUND: Idiopathic rapid eye movement sleep behavior disorder (iRBD) represents the prodromal stage of α-synucleinopathies. Reliable biomarkers are needed to predict phenoconversion. OBJECTIVE: The aim was to derive and validate a brain glucose metabolism pattern related to phenoconversion in iRBD (iRBDconvRP) using spatial covariance analysis (Scaled Subprofile Model and Principal Component Analysis [SSM-PCA]). METHODS: Seventy-six consecutive iRBD patients (70 ± 6 years, 15 women) were enrolled in two centers and prospectively evaluated to assess phenoconversion (30 converters, 73 ± 6 years, 14 Parkinson's disease and 16 dementia with Lewy bodies, follow-up time: 21 ± 14 months; 46 nonconverters, 69 ± 6 years, follow-up time: 33 ± 19 months). All patients underwent [18 F]FDG-PET (18 F-fluorodeoxyglucose positron emitting tomography) to investigate brain glucose metabolism at baseline. SSM-PCA was applied to obtain the iRBDconvRP; nonconverter patients were considered as the reference group. Survival analysis and Cox regression were applied to explore prediction power. RESULTS: First, we derived and validated two distinct center-specific iRBDconvRP that were comparable and significantly able to predict phenoconversion. Then, SSM-PCA was applied to the whole set, identifying the iRBDconvRP. The iRBDconvRP included positive voxel weights in cerebellum; brainstem; anterior cingulate cortex; lentiform nucleus; and middle, mesial temporal, and postcentral areas. Negative voxel weights were found in posterior cingulate, precuneus, middle frontal gyrus, and parietal areas. Receiver operating characteristic analysis showed an area under the curve of 0.85 (sensitivity: 87%, specificity: 72%), discriminating converters from nonconverters. The iRBDconvRP significantly predicted phenoconversion (hazard ratio: 7.42, 95% confidence interval: 2.6-21.4). CONCLUSIONS: We derived and validated an iRBDconvRP to efficiently discriminate converter from nonconverter iRBD patients. [18 F]FDG-PET pattern analysis has potential as a phenoconversion biomarker in iRBD patients. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Sujet(s)
Maladie de Parkinson , Trouble du comportement en sommeil paradoxal , Humains , Femelle , Fluorodésoxyglucose F18 , Sommeil paradoxal , Trouble du comportement en sommeil paradoxal/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/métabolisme , Marqueurs biologiques , Glucose/métabolisme
13.
Eur J Neurol ; 29(12): 3590-3599, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-36047985

RÉSUMÉ

BACKGROUND AND PURPOSE: The insidious onset of Parkinson's disease (PD) makes early diagnosis difficult. Notably, idiopathic rapid eye movement sleep behavior disorder (iRBD) was reported as a prodrome of PD, which may represent a breakthrough for the early diagnosis of PD. However, currently there is no reliable biomarker for PD diagnosis. Considering that α-synuclein (α-Syn) and neuroinflammation are known to develop prior to the onset of clinical symptoms in PD, it was hypothesized that plasma total exosomal α-Syn (t-exo α-Syn), neural-derived exosomal α-Syn (n-exo α-Syn) and exosomal apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC) may be potential biomarkers of PD. METHODS: In this study, 78 PD patients, 153 probable iRBD patients (pRBD) and 63 healthy controls (HCs) were recruited. α-Syn concentrations were measured using a one-step paramagnetic particle-based chemiluminescence immunoassay, and ASC levels were measured using the Ella system. RESULTS: It was found that t-exo α-Syn was significantly increased in the PD group compared to the pRBD and HC groups (p < 0.0001), whilst n-exo α-Syn levels were significantly increased in both the PD and pRBD groups compared to HCs (p < 0.0001). Furthermore, although no difference was found in ASC levels between the PD and pRBD groups, there was a positive correlation between ASC and α-Syn in exosomes. CONCLUSIONS: Our results suggest that both t-exo α-Syn and n-exo α-Syn were elevated in the PD group, whilst only n-exo α-Syn was elevated in the pRBD group. Additionally, the adaptor protein of inflammasome ASC is correlated with α-Syn and may facilitate synucleinopathy.


Sujet(s)
Exosomes , Maladie de Parkinson , Trouble du comportement en sommeil paradoxal , Humains , Trouble du comportement en sommeil paradoxal/métabolisme , alpha-Synucléine , Maladie de Parkinson/diagnostic , Exosomes/métabolisme , Marqueurs biologiques
14.
Comput Methods Programs Biomed ; 225: 107042, 2022 Oct.
Article de Anglais | MEDLINE | ID: mdl-35970056

RÉSUMÉ

BACKGROUND AND OBJECTIVES: 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) combined with principal component analysis (PCA) has been applied to identify disease-related brain patterns in neurodegenerative disorders such as Parkinson's disease (PD), Dementia with Lewy Bodies (DLB) and Alzheimer's disease (AD). These patterns are used to quantify functional brain changes at the single subject level. This is especially relevant in determining disease progression in idiopathic REM sleep behavior disorder (iRBD), a prodromal stage of PD and DLB. However, the PCA method is limited in discriminating between neurodegenerative conditions. More advanced machine learning algorithms may provide a solution. In this study, we apply Generalized Matrix Learning Vector Quantization (GMLVQ) to FDG-PET scans of healthy controls, and patients with AD, PD and DLB. Scans of iRBD patients, scanned twice with an approximate 4 year interval, were projected into GMLVQ space to visualize their trajectory. METHODS: We applied a combination of SSM/PCA and GMLVQ as a classifier on FDG-PET data of healthy controls, AD, DLB, and PD patients. We determined the diagnostic performance by performing a ten times repeated ten fold cross validation. We analyzed the validity of the classification system by inspecting the GMLVQ space. First by the projection of the patients into this space. Second by representing the axis, that span this decision space, into a voxel map. Furthermore, we projected a cohort of RBD patients, whom have been scanned twice (approximately 4 years apart), into the same decision space and visualized their trajectories. RESULTS: The GMLVQ prototypes, relevance diagonal, and decision space voxel maps showed metabolic patterns that agree with previously identified disease-related brain patterns. The GMLVQ decision space showed a plausible quantification of FDG-PET data. Distance traveled by iRBD subjects through GMLVQ space per year (i.e. velocity) was correlated with the change in motor symptoms per year (Spearman's rho =0.62, P=0.004). CONCLUSION: In this proof-of-concept study, we show that GMLVQ provides a classification of patients with neurodegenerative disorders, and may be useful in future studies investigating speed of progression in prodromal disease stages.


Sujet(s)
Maladies neurodégénératives , Maladie de Parkinson , Trouble du comportement en sommeil paradoxal , Fluorodésoxyglucose F18 , Humains , Maladies neurodégénératives/imagerie diagnostique , Maladie de Parkinson/imagerie diagnostique , Tomographie par émission de positons/méthodes , Trouble du comportement en sommeil paradoxal/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/métabolisme
16.
J Neurosci Res ; 100(10): 1815-1833, 2022 10.
Article de Anglais | MEDLINE | ID: mdl-35790021

RÉSUMÉ

Rapid eye movement (REM) sleep behavior disorder (RBD) is a parasomnia characterized by polysomnography-confirmed REM sleep without atonia and dream-enacting behaviors. This disorder is considered a prodromal syndrome of alpha-synucleinopathies like Parkinson's disease (PD), where it affects more than 50% of PD patients. The underlying pathology of RBD has been generally understood to involve the pontine nuclei within the brainstem. However, the complete pathophysiology beyond the brainstem remains unclear as does its relationship with PD pathology. Therefore, this review aims to survey the neuroimaging literature involving PET, SPECT, and MR imaging techniques to provide an updated understanding of the neuro-chemical, structural, and functional changes in both RBD and PD patients comorbid with RBD. This review found neuroimaging evidence that indicate alterations to the dopaminergic and cholinergic system, blood perfusion, and glucose metabolism in both RBD patients and PD patients with RBD. Beyond the brainstem, structural and functional changes were found to involve the nigrostriatal system, limbic system, and the cortex-suggesting that RBD is a multi-systemic neurodegenerative process. Future investigations are encouraged to follow RBD patients longitudinally using multimodal imaging techniques to enhance our understanding of this parasomnia disorder. Uncovering which individuals are most likely to develop an alpha-synuclein disorder in the prodromal phase will improve patient outcomes and potentially aid in the development of novel treatments for patients affected by RBD.


Sujet(s)
Maladie de Parkinson , Trouble du comportement en sommeil paradoxal , Synucléinopathies , Humains , Neuroimagerie , Maladie de Parkinson/complications , Maladie de Parkinson/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/métabolisme , Sommeil paradoxal/physiologie , Synucléinopathies/imagerie diagnostique
17.
Brain ; 145(9): 3162-3178, 2022 09 14.
Article de Anglais | MEDLINE | ID: mdl-35594873

RÉSUMÉ

Isolated REM sleep behaviour disorder (iRBD) is a synucleinopathy characterized by abnormal behaviours and vocalizations during REM sleep. Most iRBD patients develop dementia with Lewy bodies, Parkinson's disease or multiple system atrophy over time. Patients with iRBD exhibit brain atrophy patterns that are reminiscent of those observed in overt synucleinopathies. However, the mechanisms linking brain atrophy to the underlying alpha-synuclein pathophysiology are poorly understood. Our objective was to investigate how the prion-like and regional vulnerability hypotheses of alpha-synuclein might explain brain atrophy in iRBD. Using a multicentric cohort of 182 polysomnography-confirmed iRBD patients who underwent T1-weighted MRI, we performed vertex-based cortical surface and deformation-based morphometry analyses to quantify brain atrophy in patients (67.8 years, 84% male) and 261 healthy controls (66.2 years, 75%) and investigated the morphological correlates of motor and cognitive functioning in iRBD. Next, we applied the agent-based Susceptible-Infected-Removed model (i.e. a computational model that simulates in silico the spread of pathologic alpha-synuclein based on structural connectivity and gene expression) and tested if it recreated atrophy in iRBD by statistically comparing simulated regional brain atrophy to the atrophy observed in patients. The impact of SNCA and GBA gene expression and brain connectivity was then evaluated by comparing the model fit to the one obtained in null models where either gene expression or connectivity was randomized. The results showed that iRBD patients present with cortical thinning and tissue deformation, which correlated with motor and cognitive functioning. Next, we found that the computational model recreated cortical thinning (r = 0.51, P = 0.0007) and tissue deformation (r = 0.52, P = 0.0005) in patients, and that the connectome's architecture along with SNCA and GBA gene expression contributed to shaping atrophy in iRBD. We further demonstrated that the full agent-based model performed better than network measures or gene expression alone in recreating the atrophy pattern in iRBD. In summary, atrophy in iRBD is extensive, correlates with motor and cognitive function and can be recreated using the dynamics of agent-based modelling, structural connectivity and gene expression. These findings support the concepts that both prion-like spread and regional susceptibility account for the atrophy observed in prodromal synucleinopathies. Therefore, the agent-based Susceptible-Infected-Removed model may be a useful tool for testing hypotheses underlying neurodegenerative diseases and new therapies aimed at slowing or stopping the spread of alpha-synuclein pathology.


Sujet(s)
Maladies neurodégénératives , Prions , Trouble du comportement en sommeil paradoxal , Synucléinopathies , Sujet âgé , Atrophie/anatomopathologie , Encéphale/anatomopathologie , Amincissement du cortex cérébral , Femelle , Expression des gènes , Humains , Mâle , Maladies neurodégénératives/anatomopathologie , Prions/métabolisme , Trouble du comportement en sommeil paradoxal/métabolisme , Synucléinopathies/imagerie diagnostique , Synucléinopathies/génétique , alpha-Synucléine/génétique , alpha-Synucléine/métabolisme
18.
Parkinsonism Relat Disord ; 95: 47-53, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35030449

RÉSUMÉ

OBJECTIVE: With a dual-tracer PET study design, this study aimed to identify the differences between PD patients with and without probable RBD (PD+RBD+/PD+RBD-) and the influence of timing of RBD onset [probable RBD anterior to PD onset (PD+RBDa)/probable RBD posterior to PD onset (PD+RBDp)]. METHODS: Seventy-four PD+RBD+ patients, sixty-three PD+RBD-patients and twenty healthy controls were enrolled. Clinical variates, striatal DAT tracer uptake, voxel-wise glucose metabolism and Parkinson's disease-related pattern (PDRP) expressions were compared among groups. RESULTS: No significant difference were found on clinical characteristics between PD+RBD+ and PD+RBD-groups. Compared with PD+RBD-group, PD+RBD+ group had more severe dopaminergic dysfunction (p<0.05) except for posterior putamen in the more affected hemisphere (MAH) (p = 0.350). Meanwhile, it showed relative hypermetabolism in anterior putamen in the less affected hemisphere (LAH), bilateral anterior pallidum with wider involvement in the LAH, hippocampus and para-hippocampus in the LAH and bilateral olfactory gyrus, together with relative hypometabolism in limited bilateral posterio-parietal area (p<0.001). Significantly elevated PDRP expression was also seen in PD+RBD+ group (p < 0.01). For the timing of RBD onset, PD+RBDa patients harbored greater progression rate than PD+RBDp patients (p<0.01), greater DAT declining rates of striatal subregions and greater increasing rate of PDRP expressions than both PD+RBDp and PD+RBD-patients (p<0.05). CONCLUSION: Our study found that PD patients with probable RBD have worse striatal dopaminergic dysfunction and higher PDRP network activity, supporting the assumption that PD with RBD may be a specific phenotype of PD. Additionally, RBD preceding PD onset may indicate a steeper disease decline.


Sujet(s)
Maladie de Parkinson , Trouble du comportement en sommeil paradoxal , Corps strié/imagerie diagnostique , Corps strié/métabolisme , Dopamine/métabolisme , Transporteurs de la dopamine/métabolisme , Humains , Maladie de Parkinson/complications , Maladie de Parkinson/imagerie diagnostique , Maladie de Parkinson/métabolisme , Tomographie par émission de positons , Trouble du comportement en sommeil paradoxal/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/étiologie , Trouble du comportement en sommeil paradoxal/métabolisme
19.
Neurobiol Dis ; 164: 105626, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35031485

RÉSUMÉ

Braak's hypothesis has been extremely influential over the last two decades. However, neuropathological and clinical evidence suggest that the model does not conform to all patients with Parkinson's disease (PD). To resolve this controversy, a new model was recently proposed; in brain-first PD, the initial α-synuclein pathology arise inside the central nervous system, likely rostral to the substantia nigra pars compacta, and spread via interconnected structures - eventually affecting the autonomic nervous system; in body-first PD, the initial pathological α-synuclein originates in the enteric nervous system with subsequent caudo-rostral propagation to the autonomic and central nervous system. By using REM-sleep behavior disorder (RBD) as a clinical identifier to distinguish between body-first PD (RBD-positive at motor symptom onset) and brain-first PD (RBD-negative at motor symptom onset), we explored the literature to evaluate clinical and imaging differences between these proposed subtypes. Body-first PD patients display: 1) a larger burden of autonomic symptoms - in particular orthostatic hypotension and constipation, 2) more frequent pathological α-synuclein in peripheral tissues, 3) more brainstem and autonomic nervous system involvement in imaging studies, 4) more symmetric striatal dopaminergic loss and motor symptoms, and 5) slightly more olfactory dysfunction. In contrast, only minor cortical metabolic alterations emerge before motor symptoms in body-first. Brain-first PD is characterized by the opposite clinical and imaging patterns. Patients with pathological LRRK2 genetic variants mostly resemble a brain-first PD profile whereas patients with GBA variants typically conform to a body-first profile. SNCA-variant carriers are equally distributed between both subtypes. Overall, the literature indicates that body-first and brain-first PD might be two distinguishable entities on some clinical and imaging markers.


Sujet(s)
Encéphale/imagerie diagnostique , Maladie de Parkinson/imagerie diagnostique , Trouble du comportement en sommeil paradoxal/imagerie diagnostique , Encéphale/métabolisme , Humains , Maladie de Parkinson/métabolisme , Trouble du comportement en sommeil paradoxal/métabolisme , alpha-Synucléine/métabolisme
20.
Pharmacol Biochem Behav ; 213: 173319, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34990706

RÉSUMÉ

We aimed to determine whether REM sleep deprivation (RSD) affects extinction and reinstatement of methamphetamine (METH) reward memory in male rats and also to evaluate the possible role of dopamine D1-like and D2-like dopamine (DA) receptors in these processes. Male rats were trained to acquire METH-induced place preference (2 mg/kg, i.p.). METH reward memory was then reinstated following a 10-day extinction period. The animals underwent a 72-hour sleep deprivation episode by multiple platforms method (in separate groups), either before the extraction or before the reinstatement of METH reward memory. The animals received SCH 23390 (0.01 or 0.05 mg/kg, i.p.) or sulpiride (20 or 60 mg/kg, i.p.) as antagonists of D1-like and D2-like DA receptors, respectively, either immediately following each daily extinction session or before the reinstatement of METH-seeking behavior. The RSD episode postponed extinction and facilitated reinstatement of METH reward memory. Administration of SCH 23390, but not sulpiride, facilitated METH extinction and decreased reinstatement of the extinguished METH-seeking behavior. Moreover, locomotor activity was not affected by METH and/or the RSD paradigm. The results would seem to suggest that the D1-like, but not the D2-like, DA receptors may be involved in the extinction and reinstatement of the extinguished METH reward memory in RSD animals. Nonetheless, more investigations are needed to elucidate the exact mechanisms involved.


Sujet(s)
Comportement de recherche de substances/effets des médicaments et des substances chimiques , Métamfétamine/pharmacologie , Trouble du comportement en sommeil paradoxal/métabolisme , Récepteur dopamine D1/métabolisme , Privation de sommeil/métabolisme , Animaux , Benzazépines/pharmacologie , Conditionnement psychologique/effets des médicaments et des substances chimiques , Agents dopaminergiques/pharmacologie , Extinction (psychologie)/effets des médicaments et des substances chimiques , Locomotion/effets des médicaments et des substances chimiques , Mâle , Mémoire/effets des médicaments et des substances chimiques , Rats , Rat Wistar , Récepteur D2 de la dopamine/métabolisme , Récompense , Sommeil paradoxal , Sulpiride/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE