Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 643
1.
Parkinsonism Relat Disord ; 122: 106947, 2024 May.
Article En | MEDLINE | ID: mdl-38547558

INTRODUCTION: Autonomic dysfunction (AuD) is a significant clinical challenge in patients with Dementia with Lewy Bodies (DLB). Manifestations of AuD such as orthostatic hypotension (OH) is associated with falls and decreased quality of life. Cardiac autonomic denervation is an early phenomenon in DLB and a potential contributor to OH. This retrospective study was undertaken to explore whether routine ECG tracings could be used to identify signs of autonomic dysfunction in DLB. METHODS: 18 patients with DLB and 18 age-matched patients with Alzheimer's disease (AD) were included. ECGs and clinical data were analyzed retrospectively for heart rate variability (HRV) and QTc interval prolongation. RESULTS: During an average of 10 years observation time (first to last ECG recording), the QTc interval increased in the DLB group, but not in the AD group. HRV was significantly lower at end of follow-up in the DLB group than in the AD group. DLB patients with OH had greater QTc prolongation. CONCLUSION: Longitudinal ECG analysis indicates that signs of AuD in DLB are reflected on routine ECG tracings. If confirmed in larger cohorts, this could influence risk stratification and help direct preventive measures.


Alzheimer Disease , Electrocardiography , Heart Rate , Lewy Body Disease , Humans , Male , Female , Aged , Alzheimer Disease/physiopathology , Lewy Body Disease/physiopathology , Lewy Body Disease/complications , Heart Rate/physiology , Retrospective Studies , Aged, 80 and over , Long QT Syndrome/physiopathology , Long QT Syndrome/etiology , Disease Progression , Hypotension, Orthostatic/etiology , Hypotension, Orthostatic/physiopathology , Middle Aged
2.
Sleep ; 47(5)2024 May 10.
Article En | MEDLINE | ID: mdl-38330231

STUDY OBJECTIVES: Isolated rapid eye movement sleep behavior disorder (iRBD) is a prodromal stage of α-synucleinopathies and eventually phenoconverts to overt neurodegenerative diseases including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Associations of baseline resting-state electroencephalography (EEG) with phenoconversion have been reported. In this study, we aimed to develop machine learning models to predict phenoconversion time and subtype using baseline EEG features in patients with iRBD. METHODS: At baseline, resting-state EEG and neurological assessments were performed on patients with iRBD. Calculated EEG features included spectral power, weighted phase lag index, and Shannon entropy. Three models were used for survival prediction, and four models were used for α-synucleinopathy subtype prediction. The models were externally validated using data from a different institution. RESULTS: A total of 236 iRBD patients were followed up for up to 8 years (mean 3.5 years), and 31 patients converted to α-synucleinopathies (16 PD, 9 DLB, 6 MSA). The best model for survival prediction was the random survival forest model with an integrated Brier score of 0.114 and a concordance index of 0.775. The K-nearest neighbor model was the best model for subtype prediction with an area under the receiver operating characteristic curve of 0.901. Slowing of the EEG was an important feature for both models. CONCLUSIONS: Machine learning models using baseline EEG features can be used to predict phenoconversion time and its subtype in patients with iRBD. Further research including large sample data from many countries is needed to make a more robust model.


Electroencephalography , Machine Learning , REM Sleep Behavior Disorder , Humans , REM Sleep Behavior Disorder/physiopathology , REM Sleep Behavior Disorder/diagnosis , Male , Female , Electroencephalography/methods , Aged , Middle Aged , Lewy Body Disease/physiopathology , Synucleinopathies/physiopathology , Disease Progression , Prodromal Symptoms
3.
CNS Neurosci Ther ; 28(2): 183-205, 2022 02.
Article En | MEDLINE | ID: mdl-34873859

AIMS: The aim of this study was to identify brain regions with local, structural, and functional abnormalities in dementia with Lewy bodies (DLB) and uncover the differences between DLB and Alzheimer's disease (AD). The neural networks involved in the identified abnormal brain regions were further described. METHODS: PubMed, Web of Science, OVID, Science Direct, and Cochrane Library databases were used to identify neuroimaging studies that included DLB versus healthy controls (HCs) or DLB versus AD. The coordinate-based meta-analysis and functional meta-analytic connectivity modeling were performed using the activation likelihood estimation algorithm. RESULTS: Eleven structural studies and fourteen functional studies were included in this quantitative meta-analysis. DLB patients showed a dysfunction in the bilateral inferior parietal lobule and right lingual gyrus compared with HC patients. DLB patients showed a relative preservation of the medial temporal lobe and a tendency of lower metabolism in the right lingual gyrus compared with AD. The frontal-parietal, salience, and visual networks were all abnormally co-activated in DLB, but the default mode network remained normally co-activated compared with AD. CONCLUSIONS: The convergence of local brain regions and co-activation neural networks might be potential specific imaging markers in the diagnosis of DLB. This might provide a pathway for the neural regulation in DLB patients, and it might contribute to the development of specific interventions for DLB and AD.


Alzheimer Disease , Lewy Body Disease , Neuroimaging , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Humans , Lewy Body Disease/diagnostic imaging , Lewy Body Disease/pathology , Lewy Body Disease/physiopathology , Likelihood Functions
4.
Science ; 374(6571): 1106-1113, 2021 Nov 26.
Article En | MEDLINE | ID: mdl-34672693

Disrupted hippocampal performance underlies psychiatric comorbidities and cognitive impairments in patients with neurodegenerative disorders. To understand the contribution of adult hippocampal neurogenesis (AHN) to amyotrophic lateral sclerosis, Huntington's disease, Parkinson's disease, dementia with Lewy bodies, and frontotemporal dementia, we studied postmortem human samples. We found that adult-born dentate granule cells showed abnormal morphological development and changes in the expression of differentiation markers. The ratio of quiescent to proliferating hippocampal neural stem cells shifted, and the homeostasis of the neurogenic niche was altered. Aging and neurodegenerative diseases reduced the phagocytic capacity of microglia, triggered astrogliosis, and altered the microvasculature of the dentate gyrus. Thus, enhanced vulnerability of AHN to neurodegeneration might underlie hippocampal dysfunction during physiological and pathological aging in humans.


Hippocampus/physiopathology , Neurodegenerative Diseases/physiopathology , Neurogenesis , Adult , Aged , Aged, 80 and over , Aging , Amyotrophic Lateral Sclerosis/physiopathology , Cell Proliferation , Dentate Gyrus/blood supply , Dentate Gyrus/pathology , Dentate Gyrus/physiopathology , Female , Frontotemporal Dementia/physiopathology , Hippocampus/pathology , Humans , Huntington Disease/physiopathology , Lewy Body Disease/physiopathology , Male , Microglia/physiology , Middle Aged , Neural Stem Cells/physiology , Neurodegenerative Diseases/pathology , Parkinson Disease/physiopathology , Phagocytosis
5.
Neurology ; 97(10): e1031-e1040, 2021 09 07.
Article En | MEDLINE | ID: mdl-34404743

OBJECTIVE: This study aimed to quantify the trajectory and magnitude of change of the key clinical features and corresponding symptom domains of dementia with Lewy bodies (DLB) and Parkinson disease dementia (PDD), including global cognition, parkinsonism, recurrent visual hallucinations, cognitive fluctuations, and sleep disturbance. METHODS: One hundred sixteen patients with Lewy body dementia (DLB = 72, PDD = 44) underwent assessment at baseline and 3 and 6 months as part of a prospective multicenter randomized controlled trial. Linear mixed models were constructed for core outcome measures using the Mini-Mental State Examination (MMSE), motor section of the Unified Parkinson's Disease Rating Scale (UPDRS-III), Dementia Cognitive Fluctuations Scale (DCFS), and Neuropsychiatric Inventory (NPI). RESULTS: Within the time frame of our study (6 months), we were able to identify a significant cognitive decline of 1.3 points on the MMSE (p = 0.002) and significant worsening of motor parkinsonism with an increase in UPDRS-III score of 3.2 points (p = 0.018). Fluctuation severity also increased using the DCFS with a 6-month change in score of 1.3 points (p = 0.001). Uniquely, a signal for increased severity of sleep symptoms of 1.2 points (NPI-sleep) was also detectable (p = 0.04). Significant changes in neuropsychiatric symptoms were not detected. There was no difference in rates of change of scores between DLB and PDD. DISCUSSION: Clinically significant rates of change in core clinical features can be detected and quantified in Lewy body dementia over a relatively short period (6 months) using common clinical instruments and thus may be useful as clinical endpoints for therapeutic trials of disease-modifying and symptomatic agents.


Disease Progression , Lewy Body Disease , Aged , Aged, 80 and over , Cognitive Dysfunction/physiopathology , Female , Humans , Lewy Body Disease/physiopathology , Longitudinal Studies , Male , Parkinson Disease/physiopathology
6.
Neurology ; 97(8): e825-e835, 2021 08 24.
Article En | MEDLINE | ID: mdl-34088871

OBJECTIVE: The associations of Lewy bodies (LBs) with olfactory dysfunction, parkinsonism, and higher odds of dementia were assessed in Black and White community-dwelling elders and racial differences in these associations were tested. METHODS: Black decedents (n = 81) were matched 2-to-1 by age, sex, years of education, and follow-up time in the study with White decedents (n = 154) from 4 longitudinal studies of dementia and aging. Participants underwent uniform clinical examination and cognitive, motor, and olfactory testing. LBs were detected in 7 brain regions by α-synuclein immunohistochemistry and racial differences in their association with olfaction, parkinsonism, and odds of dementia were determined using regression analyses. RESULTS: The mean scores of the odor test, global parkinsonism signs, and global cognition were lower in Black than White decedents; the frequency of dementia was similar in both groups. The frequency of LBs was similar in Black and White decedents (∼25%), as was the frequency of LBs in individual brain regions, while the mean LB counts/mm2 were similar in all regions except the cingulate cortex, which showed higher mean LB counts in Black decedents. In regression analyses, LBs were associated with impaired olfaction (-2.23, 95% confidence interval [CI] -3.45 to -1.01) and higher odds of dementia (odds ratio 3.0, 95% CI 1.10-8.17) in both racial groups; an association with parkinsonism was stronger in Black than White decedents. CONCLUSIONS: The frequency, distribution, and clinical manifestations of LBs are similar in Black and White elders.


Amygdala/pathology , Black or African American/ethnology , Cerebral Cortex/pathology , Lewy Bodies/pathology , Lewy Body Disease/ethnology , Olfaction Disorders/ethnology , Substantia Nigra/pathology , White People/ethnology , Aged , Aged, 80 and over , Autopsy , Female , Humans , Lewy Body Disease/complications , Lewy Body Disease/pathology , Lewy Body Disease/physiopathology , Longitudinal Studies , Male , Olfaction Disorders/etiology , Olfaction Disorders/physiopathology , Severity of Illness Index , United States/ethnology
7.
Neuropsychology ; 35(3): 276-284, 2021 Mar.
Article En | MEDLINE | ID: mdl-33970661

Objectives: In dementia with Lewy bodies (DLB) recurrent visual hallucinations (VH) often coexist or occur consecutively to impaired visual perception. Since in-depth neuropsychological testing is time-consuming, and therefore, not routinely performed in clinical settings, we aimed to explore whether standard cognitive screening tests may be helpful to alert a clinician to the presence of VH in DLB by exploring association between visuo-spatial dysfunction and VH. Method: The clock drawing, cube, and pentagons copying items from Montreal Cognitive Assessment and Mini-Mental State Exam and nonmotor Hooper visual organization test (HVOT) have been scored in DLB patients with and without VH using traditional and extended scoring methods. Results: Forty-five of 69 (65%) DLB patients were VH-positive (VH+). VH+ patients performed worse on the clock drawing (8.8/16 vs. 11.9/16, p = .016) with a higher rate of misrepresentation of time (69% vs. 29%, p = .002) and numbers (53% vs. 25%, p = .024). Likewise, VH+ patients performed worse on the HVOT (13.3/30 vs. 15.7/30, p = .009) having more isolated (6.2/30 vs. 4.4/30, p = .012) types of responses compared to VH- patients. Both groups had similar copying ability (p > .05). The VH discriminative accuracy of the clock drawing was comparable to that of the more elaborate test of visual perception, the HVOT. Conclusions: In DLB impaired drawing and visual organization, but not copying ability is associated with the presence of VH. The simple clock drawing test can be helpful to alert a clinician to the possibility of VH in DLB. (PsycInfo Database Record (c) 2021 APA, all rights reserved).


Hallucinations/physiopathology , Lewy Body Disease/physiopathology , Spatial Processing , Aged , Aged, 80 and over , Female , Hallucinations/psychology , Humans , Lewy Body Disease/psychology , Male , Neuropsychological Tests , Spatial Navigation , Visual Perception
8.
Neurology ; 96(23): e2801-e2811, 2021 06 08.
Article En | MEDLINE | ID: mdl-33883238

OBJECTIVE: To provide evidence that cardiac I-123-metaiodobenzylguanidine sympathetic innervation imaging (MIBG) scintigraphy differentiates probable mild cognitive impairment with Lewy bodies (MCI-LB) from mild cognitive impairment due to Alzheimer disease (MCI-AD), we scanned patients with MCI and obtained consensus clinical diagnoses of their MCI subtype. We also performed baseline FP-CIT scans to compare the accuracy of MIBG and FP-CIT. METHODS: We conducted a prospective cohort study into the accuracy of cardiac MIBG scintigraphy in the diagnosis of MCI-LB. Follow-up clinical assessment was used to diagnose MCI-AD (no core features of MCI-LB and normal FP-CIT), probable MCI-LB (2 or more core features, or 1 core feature with abnormal FP-CIT), or possible MCI-LB (1 core feature or abnormal FP-CIT). For the comparison between MIBG and FP-CIT, only core clinical features were used for diagnosis. RESULTS: We recruited 95 people with mild cognitive impairment. Cardiac MIBG was abnormal in 22/37 probable and 2/15 possible MCI-LB cases and normal in 38/43 MCI-AD cases. The sensitivity in probable MCI-LB was 59% (95% confidence interval [CI], 42%-75%), specificity 88% (75%-96%), and accuracy 75% (64%-84%). The positive likelihood ratio was 5.1 and negative likelihood ratio 0.46. With symptom-only diagnoses, the accuracies were 79% for MIBG (95% CI, 68%-87%) and 76% for FP-CIT (95% CI, 65%-85%). CONCLUSIONS: Cardiac MIBG appears useful in early disease, with an abnormal scan highly suggestive of MCI-LB. Validation in a multicenter setting is justified. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that cardiac MIBG distinguishes MCI-LB from MCI-AD.


Alzheimer Disease/diagnostic imaging , Cognitive Dysfunction/diagnostic imaging , Lewy Body Disease/diagnostic imaging , Myocardial Perfusion Imaging/standards , Tomography, Emission-Computed, Single-Photon/standards , 3-Iodobenzylguanidine , Aged , Aged, 80 and over , Alzheimer Disease/complications , Alzheimer Disease/physiopathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Diagnosis, Differential , Female , Follow-Up Studies , Heart/innervation , Humans , Lewy Body Disease/complications , Lewy Body Disease/physiopathology , Male , Sensitivity and Specificity , Tropanes
9.
Brain Res ; 1765: 147481, 2021 08 15.
Article En | MEDLINE | ID: mdl-33865805

Amino acids' neuroactivity, and roles in excitotoxity and oxidative stress are linked to dementia. We aimed to investigate whether circulating amino acid concentrations were associated with cognitive decline in patients with mild Alzheimer's disease (AD) and Lewy body dementia (LBD). Baseline serum amino acid concentrations were measured in 89 patients with AD and 65 with LBD (13 with Parkinson's disease dementia and 52 with dementia with Lewy bodies). The Mini-Mental State Examination (MMSE) was administered at baseline and annually for five years. Associations between baseline amino acid concentrations and longitudinal MMSE score were assessed using a linear-mixed effects model stratified by diagnosis with adjustment for multiple comparisons. The results of the study indicated that serum tyrosine was positively associated with MMSE performance during the five-year follow-up period in patients with LBD (q-value = 0.012), but not AD. In conclusion, higher baseline serum concentrations of tyrosine, the precursor amino acid in dopamine and norepinephrine synthesis, was associated with better cognitive performance in patients with LBD, but not AD, throughout the 5-year follow-up period.


Cognition/physiology , Lewy Body Disease/metabolism , Tyrosine/analysis , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Biomarkers/blood , Cognitive Dysfunction/metabolism , Cohort Studies , Female , Humans , Lewy Body Disease/physiopathology , Male , Tyrosine/blood , Tyrosine/metabolism
10.
Expert Opin Investig Drugs ; 30(6): 603-609, 2021 Jun.
Article En | MEDLINE | ID: mdl-33899637

Introduction: Dementia with Lewy bodies (DLB) is characterized by the toxic accumulation of α-synuclein protein inside neural cells; this results in neurodegeneration which is clinically accompanied by behavioral and psychological changes. DLB shares features with Parkinson's disease (PD) and Parkinson's disease dementia (PDD), but also overlaps neurochemically and pathologically with Alzheimer's disease. Symptomatic treatments for LBD differ in their effectiveness while disease-modifying and curative approaches are much needed.Areas covered: We explore emerging therapeutics for DLB through the lens of repurposing approved drugs and survey their potential for disease modifying actions in DLB. Given the complexity of DLB with multiple pathologies, potential therapeutic targets that could affect Lewy body pathology, or metabolism or neurotransmitters or immunomodulation were surveyed. We queried PubMed and ClinicalTrials.gov searches 2017-2020.Expert opinion: DLB is not simply aredux ofAD or PD; hence, treatments should not be exclusively duplicative ofAD or PD directed treatments. This opens amyriad of possibilities for therapeutic approaches that are disease specific or repurposed.


Drug Development , Lewy Body Disease/drug therapy , Molecular Targeted Therapy , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Animals , Drug Repositioning , Humans , Lewy Body Disease/physiopathology , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology
11.
Neural Plast ; 2021: 8834645, 2021.
Article En | MEDLINE | ID: mdl-33854544

Neural plasticity-the ability to alter a neuronal response to environmental stimuli-is an important factor in learning and memory. Short-term synaptic plasticity and long-term synaptic plasticity, including long-term potentiation and long-term depression, are the most-characterized models of learning and memory at the molecular and cellular level. These processes are often disrupted by neurodegeneration-induced dementias. Alzheimer's disease (AD) accounts for 50% of cases of dementia. Vascular dementia (VaD), Parkinson's disease dementia (PDD), dementia with Lewy bodies (DLB), and frontotemporal dementia (FTD) constitute much of the remaining cases. While vascular lesions are the principal cause of VaD, neurodegenerative processes have been established as etiological agents of many dementia diseases. Chief among such processes is the deposition of pathological protein aggregates in vivo including ß-amyloid deposition in AD, the formation of neurofibrillary tangles in AD and FTD, and the accumulation of Lewy bodies composed of α-synuclein aggregates in DLB and PDD. The main symptoms of dementia are cognitive decline and memory and learning impairment. Nonetheless, accurate diagnoses of neurodegenerative diseases can be difficult due to overlapping clinical symptoms and the diverse locations of cortical lesions. Still, new neuroimaging and molecular biomarkers have improved clinicians' diagnostic capabilities in the context of dementia and may lead to the development of more effective treatments. Both genetic and environmental factors may lead to the aggregation of pathological proteins and altered levels of cytokines, such that can trigger the formation of proinflammatory immunological phenotypes. This cascade of pathological changes provides fertile ground for the development of neural plasticity disorders and dementias. Available pharmacotherapy and disease-modifying therapies currently in clinical trials may modulate synaptic plasticity to mitigate the effects neuropathological changes have on cognitive function, memory, and learning. In this article, we review the neural plasticity changes seen in common neurodegenerative diseases from pathophysiological and clinical points of view and highlight potential molecular targets of disease-modifying therapies.


Dementia/diagnostic imaging , Dementia/physiopathology , Neuronal Plasticity/physiology , Neurons/physiology , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Biomarkers/metabolism , Brain Diseases/diagnostic imaging , Brain Diseases/metabolism , Brain Diseases/physiopathology , Dementia/metabolism , Humans , Lewy Body Disease/diagnostic imaging , Lewy Body Disease/metabolism , Lewy Body Disease/physiopathology , alpha-Synuclein/metabolism , tau Proteins/metabolism
12.
Gait Posture ; 86: 186-191, 2021 05.
Article En | MEDLINE | ID: mdl-33756407

BACKGROUND: Treadmill training may improve gait disorders associated with neurodegenerative diseases. In Parkinson's disease (PD), treadmill training alters gait patterns after one session, and long-term training improves gait parameters, fall risk, and quality of life. RESEARCH QUESTION: What is the feasibility and safety of using this intervention for people with Lewy body dementia (LBD) or Huntington's disease (HD)? METHODS: In this observational study, 10 individuals with HD, 8 individuals with LBD, and 10 control individuals walked for 20 min on a treadmill using a speed dependent protocol starting at a slow comfortable speed and increasing incrementally toward their normal overground speed. Feasibility was determined by compliance to protocol and safety by no incidents of abnormal vital signs or expressions of distress. Changes in gait measures, Timed Up and Go (TUG) scores and quantitative motor function measures (Q-Motor; precision grasp force variability, finger and foot tapping frequency) before and after treadmill walking were analyzed using linear models. RESULTS: Treadmill training is feasible and safe in LBD and HD; although, participants could not initiate treadmill walking at their comfortable overground speeds, and only 3 participants with HD were able to achieve their overground walking speed within the 20-minute session. No changes in gait measures, TUG times, and Q-Motor measures were found among LBD and HD participants after treadmill walking, although control participants demonstrated significant increases in several gait measures, and foot tap frequency (estimated difference = 0.290; p = 0.026). SIGNIFICANCE: Longer and more frequent treadmill sessions may be needed to see gait and motor function effects in LBD and HD. Motor and cognitive impairments associated with these diseases may make them less amenable to the effects of treadmill training.


Exercise Therapy , Gait Disorders, Neurologic/therapy , Huntington Disease/physiopathology , Lewy Body Disease/physiopathology , Walking/physiology , Aged , Aged, 80 and over , Feasibility Studies , Female , Humans , Male , Middle Aged , Pilot Projects , Treatment Outcome
13.
Acta Neuropathol Commun ; 9(1): 46, 2021 03 20.
Article En | MEDLINE | ID: mdl-33743820

Alpha-synuclein (α-syn) aggregation is the hallmark pathological lesion in brains of patients with Parkinson's disease (PD) and related neurological disorders characterized as synucleinopathies. Accumulating evidence now indicates that α-syn deposition is also present within the gut and other peripheral organs outside the central nervous system (CNS). In the current study, we demonstrate for the first time that α-syn pathology also accumulates within the liver, the main organ responsible for substance clearance and detoxification. We further demonstrate that cultured human hepatocytes readily internalize oligomeric α-syn assemblies mediated, at least in part, by the gap junction protein connexin-32 (Cx32). Moreover, we identified a time-dependent accumulation of α-syn within the liver of three different transgenic (tg) mouse models expressing human α-syn under CNS-specific promoters, despite the lack of α-syn mRNA expression within the liver. Such a brain-to-liver transmission route could be further corroborated by detection of α-syn pathology within the liver of wild type mice one month after a single striatal α-syn injection. In contrast to the synucleinopathy models, aged mice modeling AD rarely show any amyloid-beta (Aß) deposition within the liver. In human post-mortem liver tissue, we identified cases with neuropathologically confirmed α-syn pathology containing α-syn within hepatocellular structures to a higher degree (75%) than control subjects without α-syn accumulation in the brain (57%). Our results reveal that α-syn accumulates within the liver and may be derived from the brain or other peripheral sources. Collectively, our findings indicate that the liver may play a role in the clearance and detoxification of pathological proteins in PD and related synucleinopathies.


Brain/metabolism , Brain/pathology , Liver/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Female , Humans , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Lewy Body Disease/physiopathology , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Parkinson Disease/physiopathology , Synucleinopathies/metabolism , Synucleinopathies/pathology , Synucleinopathies/physiopathology
14.
Alzheimers Dement ; 17(8): 1317-1328, 2021 08.
Article En | MEDLINE | ID: mdl-33590967

INTRODUCTION: Gait impairment is common in neurodegenerative disorders. Specifically, gait variability-the stride-to-stride fluctuations in distance and time-has been associated with neurodegeneration and cognitive impairment. However, quantitative comparisons of gait impairments across the cognitive spectrum of dementias have not been systematically investigated. METHODS: Older adults (N = 500) with subjective cognitive impairment, Parkinson disease (PD), mild cognitive impairment (MCI), PD-MCI, Alzheimer's disease (AD), PD-dementia, Lewy body dementia, and frontotemporal dementia, as well cognitive normal controls, who were assessed for their gait and cognitive performance. RESULTS: Factor analyses grouped 11 quantitative gait parameters and identified four independent gait domains: rhythm, pace, variability, and postural control, for group comparisons and classification analysis. Among these domains, only high gait variability was associated with lower cognitive performance and accurately discriminated AD from other neurodegenerative and cognitive conditions. DISCUSSION: Our findings indicate that high gait variability is a marker of cognitive-cortical dysfunction, which can help to identify Alzheimer's disease dementia.


Cognition Disorders/physiopathology , Dementia/physiopathology , Gait/physiology , Aged , Aging/physiology , Alzheimer Disease/physiopathology , Biomarkers , Brain/physiopathology , Canada , Frontotemporal Dementia/physiopathology , Humans , Lewy Body Disease/physiopathology , Parkinson Disease/physiopathology
15.
Brain ; 144(3): 781-788, 2021 04 12.
Article En | MEDLINE | ID: mdl-33521808

Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB) are related conditions that are associated with cholinergic system dysfunction. Dysfunction of the nucleus basalis of Meynert (NBM), a basal forebrain structure that provides the dominant source of cortical cholinergic innervation, has been implicated in the pathogenesis of both PDD and DLB. Here we leverage the temporal resolution of magnetoencephalography with the spatial resolution of MRI tractography to explore the intersection of functional and structural connectivity of the NBM in a unique cohort of PDD and DLB patients undergoing deep brain stimulation of this structure. We observe that NBM-cortical structural and functional connectivity correlate within spatially and spectrally segregated networks including: (i) a beta band network to supplementary motor area, where activity in this region was found to drive activity in the NBM; (ii) a delta/theta band network to medial temporal lobe structures encompassing the parahippocampal gyrus; and (iii) a delta/theta band network to visual areas including lingual gyrus. These findings reveal functional networks of the NBM that are likely to subserve important roles in motor control, memory and visual function, respectively. Furthermore, they motivate future studies aimed at disentangling network contribution to disease phenotype.


Basal Nucleus of Meynert/physiopathology , Cerebral Cortex/physiopathology , Lewy Body Disease/physiopathology , Neural Pathways/physiopathology , Parkinson Disease/physiopathology , Deep Brain Stimulation , Diffusion Tensor Imaging , Humans , Magnetoencephalography , Nerve Net/physiopathology
16.
J Integr Neurosci ; 20(4): 1067-1078, 2021 Dec 30.
Article En | MEDLINE | ID: mdl-34997730

Parkinson's disease-related pain has increasingly been investigated in research studies. Still, only a few studies have addressed the prevalence and clinical characteristics of pain in neurodegenerative disorders with atypical parkinsonism. The existing evidence, although scarce, suggests that, similarly as in Parkinson's disease, individuals with neurodegenerative diseases with atypical parkinsonism might be predisposed to the development of persistent pain. Today, as the global population is aging and we face an epidemic of neurodegenerative disorders, under-treated pain is taking a great toll on an ever-rising number of people. Here, we provide an up-to-date review of the current knowledge on the prevalence of pain, its clinical features, and findings from experimental studies that might signpost altered pain processing in the most prevalent neurodegenerative disorders with atypical parkinsonism: multiple system atrophy, progressive supranuclear palsy, corticobasal syndrome, frontotemporal dementia, and dementia with Lewy bodies. Finally, we point out the current gaps and unmet needs that future research studies should focus on. Large-scale, high-quality clinical trials, coupled with pre-clinical research, are urgently needed to reveal the exact pathophysiological mechanisms underpinning heightened pain and pave the path for mechanistically-driven analgesic interventions to be developed, ultimately leading to an improvement in the quality of life of individuals with neurodegenerative disorders.


Corticobasal Degeneration , Frontotemporal Dementia , Lewy Body Disease , Multiple System Atrophy , Musculoskeletal Pain , Neuralgia , Supranuclear Palsy, Progressive , Corticobasal Degeneration/complications , Corticobasal Degeneration/epidemiology , Corticobasal Degeneration/physiopathology , Frontotemporal Dementia/complications , Frontotemporal Dementia/epidemiology , Frontotemporal Dementia/physiopathology , Humans , Lewy Body Disease/complications , Lewy Body Disease/epidemiology , Lewy Body Disease/physiopathology , Multiple System Atrophy/complications , Multiple System Atrophy/epidemiology , Multiple System Atrophy/physiopathology , Musculoskeletal Pain/epidemiology , Musculoskeletal Pain/etiology , Musculoskeletal Pain/physiopathology , Neuralgia/epidemiology , Neuralgia/etiology , Neuralgia/physiopathology , Prevalence , Supranuclear Palsy, Progressive/complications , Supranuclear Palsy, Progressive/epidemiology , Supranuclear Palsy, Progressive/physiopathology
17.
Am J Geriatr Psychiatry ; 29(3): 272-284, 2021 03.
Article En | MEDLINE | ID: mdl-32863138

OBJECTIVE: We explored whether the mild cognitive impairment (MCI) stages of dementia with Lewy bodies (DLB) and Alzheimer disease (AD) differ in their cognitive profiles, and longitudinal progression. DESIGN: A prospective, longitudinal design was utilized with annual follow-up (Max 5 years, Mean 1.9, standard deviation 1.1) after diagnosis. Participants underwent repeated cognitive testing, and review of their clinical diagnosis and symptoms, including evaluation of core features of DLB. SETTING: This was an observational study of independently living individuals, recruited from local healthcare trusts in North East England, UK. PARTICIPANTS: An MCI cohort (n = 76) aged ≥60 years was utilized, differentially diagnosed with MCI due to AD (MCI-AD), or possible/probable MCI with Lewy bodies (MCI-LB). MEASUREMENTS: A comprehensive clinical and neuropsychological testing battery was administered, including ACE-R, trailmaking tests, FAS verbal fluency, and computerized battery of attention and perception tasks. RESULTS: Probable MCI-LB presented with less impaired recognition memory than MCI-AD, greater initial impairments in verbal fluency and perception of line orientation, and thereafter demonstrated an expedited decline in visuo-constructional functions in the ACE-R compared to MCI-AD. No clear diagnostic group differences were found in deterioration speeds for global cognition, language, overall memory, attention or other executive functions. CONCLUSION: These findings provide further evidence for differences in severity and decline of visuospatial dysfunctions in DLB compared with AD; further exploration is required to clarify when and how differences in attention, executive, and memory functions emerge, as well as speed of decline to dementia.


Alzheimer Disease/complications , Alzheimer Disease/psychology , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/etiology , Lewy Body Disease/complications , Lewy Body Disease/psychology , Aged , Alzheimer Disease/physiopathology , Attention , Cognitive Dysfunction/physiopathology , England , Executive Function , Female , Humans , Lewy Body Disease/physiopathology , Male , Memory , Middle Aged , Neuropsychological Tests , Prospective Studies
18.
Neurology ; 96(5): e684-e697, 2021 02 02.
Article En | MEDLINE | ID: mdl-33199437

OBJECTIVES: Nucleus basalis of Meynert deep brain stimulation (NBM-DBS) has been proposed for patients with dementia. Here, we aim to assess the safety and effects of NBM-DBS in patients with Lewy body dementia (LBD), in a randomized, double-blind, crossover clinical trial. METHODS: Six patients with mild to moderate LBD (mean [SD] age, 62.2 [7.8] years) were included, operated on for bilateral NBM-DBS, and assigned to receive either active or sham NBM-DBS followed by the opposite condition for 3 months. The primary outcome was the difference in the total free recalls of the Free and Cued Selective Reminding Test (FCSRT) between active and sham NBM-DBS. Secondary outcomes were assessments of the safety and effects of NBM-DBS on cognition, motor disability, sleep, and PET imaging. RESULTS: There was no significant difference in the FCSRT score with active vs sham NBM-DBS. The surgical procedures were well tolerated in all patients, but we observed significant decreases in Stroop and Benton scores after electrode implantation. We observed no significant difference in other scales between active and sham NBM-DBS. With active NBM-DBS relative to baseline, phonemic fluency and motor disability significantly decreased. Lastly, the superior lingual gyrus metabolic activity significantly increased with active NBM-DBS. CONCLUSIONS: NBM-DBS does not appear to be totally safe for patients with LBD with no evidence of cognitive benefit. CLINICALTRIALSGOV IDENTIFIER: NCT01340001. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence that, for patients with LBD operated on for bilateral NBM-DBS, active NBM-DBS stimulation compared to sham stimulation did not significantly change selective recall scores.


Basal Nucleus of Meynert , Deep Brain Stimulation/methods , Lewy Body Disease/therapy , Mental Recall , Aged , Brain/diagnostic imaging , Cross-Over Studies , Double-Blind Method , Fluorodeoxyglucose F18 , Humans , Implantable Neurostimulators , Lewy Body Disease/physiopathology , Lewy Body Disease/psychology , Male , Middle Aged , Neuropsychological Tests , Positron-Emission Tomography , Postoperative Complications , Prosthesis Implantation , Radiopharmaceuticals , Sleep , Treatment Outcome
19.
J Alzheimers Dis ; 79(1): 59-70, 2021.
Article En | MEDLINE | ID: mdl-33216030

BACKGROUND: Plasma NfL (pNfL) levels are elevated in many neurological disorders. However, the utility of pNfL in a clinical setting has not been established. OBJECTIVE: In a cohort of diverse older participants, we examined: 1) the association of pNfL to age, sex, Hispanic ethnicity, diagnosis, and structural and amyloid imaging biomarkers; and 2) its association to baseline and longitudinal cognitive and functional performance. METHODS: 309 subjects were classified at baseline as cognitively normal (CN) or with cognitive impairment. Most subjects had structural MRI and amyloid PET scans. The most frequent etiological diagnosis was Alzheimer's disease (AD), but other neurological and neuropsychiatric disorders were also represented. We assessed the relationship of pNfL to cognitive and functional status, primary etiology, imaging biomarkers, and to cognitive and functional decline. RESULTS: pNfL increased with age, degree of hippocampal atrophy, and amyloid load, and was higher in females among CN subjects, but was not associated with Hispanic ethnicity. Compared to CN subjects, pNfL was elevated among those with AD or FTLD, but not those with neuropsychiatric or other disorders. Hippocampal atrophy, amyloid positivity and higher pNfL levels each added unique variance in predicting greater functional impairment on the CDR-SB at baseline. Higher baseline pNfL levels also predicted greater cognitive and functional decline after accounting for hippocampal atrophy and memory scores at baseline. CONCLUSION: pNfL may have a complementary and supportive role to brain imaging and cognitive testing in a memory disorder evaluation, although its diagnostic sensitivity and specificity as a stand-alone measure is modest. In the absence of expensive neuroimaging tests, pNfL could be used for differentiating neurodegenerative disease from neuropsychiatric disorders.


Alzheimer Disease/blood , Brain/diagnostic imaging , Cognitive Dysfunction/blood , Functional Status , Neurofilament Proteins/blood , Black or African American , Age Factors , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Atrophy , Brain/metabolism , Brain/pathology , Cognition , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/physiopathology , Cohort Studies , Dementia, Vascular/blood , Dementia, Vascular/diagnostic imaging , Dementia, Vascular/physiopathology , Female , Frontotemporal Lobar Degeneration/blood , Frontotemporal Lobar Degeneration/diagnostic imaging , Frontotemporal Lobar Degeneration/physiopathology , Hippocampus/diagnostic imaging , Hippocampus/pathology , Hispanic or Latino , Humans , Lewy Body Disease/blood , Lewy Body Disease/diagnostic imaging , Lewy Body Disease/physiopathology , Magnetic Resonance Imaging , Male , Middle Aged , Positron-Emission Tomography , Sex Factors , White People
20.
Ann Neurol ; 89(2): 341-357, 2021 02.
Article En | MEDLINE | ID: mdl-33217037

OBJECTIVE: Isolated (or idiopathic) rapid eye movement sleep behavior disorder (iRBD) is associated with dementia with Lewy bodies (DLB) and Parkinson's disease (PD). Biomarkers are lacking to predict conversion to a dementia or a motor-first phenotype. Here, we aimed at identifying a brain-clinical signature that predicts dementia in iRBD. METHODS: A brain-clinical signature was identified in 48 patients with polysomnography-confirmed iRBD using partial least squares between brain deformation and 27 clinical variables. The resulting variable was applied to 78 patients with iRBD followed longitudinally to predict conversion to a synucleinopathy, specifically DLB. The deformation scores from patients with iRBD were compared with 207 patients with PD, DLB, or prodromal DLB to assess if scores were higher in DLB compared to PD. RESULTS: One latent variable explained 31% of the brain-clinical covariance in iRBD, combining cortical and subcortical deformation and subarachnoid/ventricular expansion to cognitive and motor variables. The deformation score of this signature predicted conversion to a synucleinopathy in iRBD (p = 0.036, odds ratio [OR] = 2.249; 95% confidence interval [CI] = 1.053-4.803), specifically to DLB (OR = 4.754; 95% CI = 1.283-17.618, p = 0.020) and not PD (p = 0.286). Patients with iRBD who developed dementia had scores similar to clinical and prodromal patients with DLB but higher scores compared with patients with PD. The deformation score also predicted cognitive performance over 1, 2, and 4 years in patients with PD. INTERPRETATION: We identified a brain-clinical signature that predicts conversion in iRBD to more severe/dementing forms of synucleinopathy. This pattern may serve as a new biomarker to optimize patient care, target risk reduction strategies, and administer neuroprotective trials. ANN NEUROL 2021;89:341-357.


Cognition , Lewy Body Disease/physiopathology , Parkinson Disease/physiopathology , REM Sleep Behavior Disorder/physiopathology , Aged , Brain/diagnostic imaging , Case-Control Studies , Female , Humans , Least-Squares Analysis , Lewy Body Disease/diagnostic imaging , Magnetic Resonance Imaging , Male , Mental Status and Dementia Tests , Middle Aged , Parkinson Disease/diagnostic imaging , Polysomnography , Prodromal Symptoms , REM Sleep Behavior Disorder/diagnostic imaging , Synucleinopathies/diagnostic imaging , Synucleinopathies/physiopathology
...