1
|
Olney KC, Rabichow BE, Wojtas AM, DeTure M, McLean PJ, Dickson DW, Chang R, Ross OA, Fryer JD. Distinct transcriptional alterations distinguish Lewy body disease from Alzheimer's disease. Brain 2025; 148:69-88. [PMID: 38916996 PMCID: PMC11706328 DOI: 10.1093/brain/awae202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 05/08/2024] [Accepted: 06/02/2024] [Indexed: 06/27/2024] Open
Abstract
Lewy body dementia and Alzheimer's disease (AD) are leading causes of cognitive impairment, characterized by distinct but overlapping neuropathological hallmarks. Lewy body disease (LBD) is characterized by α-synuclein aggregates in the form of Lewy bodies as well as the deposition of extracellular amyloid plaques, with many cases also exhibiting neurofibrillary tangle (NFT) pathology. In contrast, AD is characterized by amyloid plaques and neurofibrillary tangles. Both conditions often co-occur with additional neuropathological changes, such as vascular disease and TDP-43 pathology. To elucidate shared and distinct molecular signatures underlying these mixed neuropathologies, we extensively analysed transcriptional changes in the anterior cingulate cortex, a brain region critically involved in cognitive processes. We performed bulk tissue RNA sequencing from the anterior cingulate cortex and determined differentially expressed genes (q-value <0.05) in control (n = 81), LBD (n = 436), AD (n = 53) and pathological amyloid cases consisting of amyloid pathology with minimal or no tau pathology (n = 39). We used gene set enrichment and weighted gene correlation network analysis to understand the pathways associated with each neuropathologically defined group. LBD cases had strong upregulation of inflammatory pathways and downregulation of metabolic pathways. The LBD cases were further subdivided into either high Thal amyloid, Braak NFT, or low pathological burden cohorts. Compared to the control cases, the LBD cohorts consistently showed upregulation for genes involved in protein folding and cytokine immune response, as well as downregulation of fatty acid metabolism. Surprisingly, concomitant tau pathology within the LBD cases resulted in no additional changes. Some core inflammatory pathways were shared between AD and LBD but with numerous disease-specific changes. Direct comparison of LBD cohorts versus AD cases revealed strong enrichment of synaptic signalling, behaviour and neuronal system pathways. Females had a stronger response overall in both LBD and AD, with several sex-specific changes. Overall, the results identify genes commonly and uniquely dysregulated in neuropathologically defined LBD and AD cases, shedding light on shared and distinct molecular pathways. Additionally, the study underscores the importance of considering sex-specific changes in understanding the complex transcriptional landscape of these neurodegenerative diseases.
Collapse
Affiliation(s)
- Kimberly C Olney
- Department of Neuroscience, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Benjamin E Rabichow
- Department of Neuroscience, Mayo Clinic, Scottsdale, AZ 85259, USA
- Program in Neuroscience, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Aleksandra M Wojtas
- Department of Neuroscience, Mayo Clinic, Scottsdale, AZ 85259, USA
- Program in Neuroscience, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Pamela J McLean
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rui Chang
- Department of Neurology, University of Arizona, Tucson, AZ 85724, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - John D Fryer
- Department of Neuroscience, Mayo Clinic, Scottsdale, AZ 85259, USA
- Program in Neuroscience, Mayo Clinic, Scottsdale, AZ 85259, USA
| |
Collapse
|
2
|
Zhu Y, Wang F, Ning P, Zhu Y, Zhang L, Li K, Liu B, Ren H, Xu Z, Pang A, Yang X. Multimodal neuroimaging-based prediction of Parkinson's disease with mild cognitive impairment using machine learning technique. NPJ Parkinsons Dis 2024; 10:218. [PMID: 39528560 PMCID: PMC11555067 DOI: 10.1038/s41531-024-00828-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
This study aimed to identify potential markers that can predict Parkinson's disease with mild cognitive impairment (PDMCI). We retrospectively collected general demographic data, clinically relevant scales, plasma samples, and neuroimaging data (T1-weighted magnetic resonance imaging (MRI) data as well as resting-state functional MRI [Rs-fMRI] data) from 173 individuals. Subsequently, based on the aforementioned multimodal indices, a support vector machine was employed to investigate the machine learning (ML) classification of PD patients with normal cognition (PDNC) and PDMCI. The performance of 29 classifiers was assessed based on various combinations of indicators. Results demonstrated that the optimal classifier in the validation set was composed by clinical + Rs-fMRI+ neurofilament light chain, exhibiting a mean Accuracy of 0.762, a mean area under curve of 0.840, a mean sensitivity of 0.745, along with a mean specificity of 0.783. The ML algorithm based on multimodal data demonstrated enhanced discriminative ability between PDNC and PDMCI patients.
Collapse
Affiliation(s)
- Yongyun Zhu
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fang Wang
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Pingping Ning
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, Shanxi, China
| | - Yangfan Zhu
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Lingfeng Zhang
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Kelu Li
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Bin Liu
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hui Ren
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhong Xu
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ailan Pang
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Xinglong Yang
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
3
|
Tullo S, Miranda AS, Del Cid-Pellitero E, Lim MP, Gallino D, Attaran A, Patel R, Novikov V, Park M, Beraldo FH, Luo W, Shlaifer I, Durcan TM, Bussey TJ, Saksida LM, Fon EA, Prado VF, Prado MAM, Chakravarty MM. Neuroanatomical and cognitive biomarkers of alpha-synuclein propagation in a mouse model of synucleinopathy prior to onset of motor symptoms. J Neurochem 2024; 168:1546-1564. [PMID: 37804203 DOI: 10.1111/jnc.15967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 10/09/2023]
Abstract
Significant evidence suggests that misfolded alpha-synuclein (aSyn), a major component of Lewy bodies, propagates in a prion-like manner contributing to disease progression in Parkinson's disease (PD) and other synucleinopathies. In fact, timed inoculation of M83 hemizygous mice with recombinant human aSyn preformed fibrils (PFF) has shown symptomatic deficits after substantial spreading of pathogenic alpha-synuclein, as detected by markers for the phosphorylation of S129 of aSyn. However, whether accumulated toxicity impact human-relevant cognitive and structural neuroanatomical measures is not fully understood. Here we performed a single unilateral striatal PFF injection in M83 hemizygous mice, and using two assays with translational potential, ex vivo magnetic resonance imaging (MRI) and touchscreen testing, we examined the combined neuroanatomical and behavioral impact of aSyn propagation. In PFF-injected mice, we observed widespread atrophy in bilateral regions that project to or receive input from the injection site using MRI. We also identified early deficits in reversal learning prior to the emergence of motor symptoms. Our findings highlight a network of regions with related cellular correlates of pathology that follow the progression of aSyn spreading, and that affect brain areas relevant for reversal learning. Our experiments suggest that M83 hemizygous mice injected with human PFF provides a model to understand how misfolded aSyn affects human-relevant pre-clinical measures and suggest that these pre-clinical biomarkers could be used to detect early toxicity of aSyn and provide better translational measures between mice and human disease.
Collapse
Affiliation(s)
- Stephanie Tullo
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Aline S Miranda
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- Departamento de Morfologia, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Esther Del Cid-Pellitero
- McGill Parkinson Program, Neurodegenerative Diseases Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Mei Peng Lim
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Daniel Gallino
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Anoosha Attaran
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Raihaan Patel
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
- Department of Biological & Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | - Vladislav Novikov
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Megan Park
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Flavio H Beraldo
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Wen Luo
- Early Drug Discovery Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Irina Shlaifer
- Early Drug Discovery Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Timothy J Bussey
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Lisa M Saksida
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Edward A Fon
- McGill Parkinson Program, Neurodegenerative Diseases Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Vania F Prado
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Marco A M Prado
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - M Mallar Chakravarty
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
- Department of Biological & Biomedical Engineering, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
4
|
Cao LX, Kong WL, Chan P, Zhang W, Morris MJ, Huang Y. Assessment tools for cognitive performance in Parkinson's disease and its genetic contributors. Front Neurol 2024; 15:1413187. [PMID: 38988604 PMCID: PMC11233456 DOI: 10.3389/fneur.2024.1413187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024] Open
Abstract
Background We have shown that genetic factors associating with motor progression of Parkinson's disease (PD), but their roles in cognitive function is poorly understood. One reason is that while cognitive performance in PD can be evaluated by various cognitive scales, there is no definitive guide indicating which tool performs better. Methods Data were obtained from the Parkinson's Progression Markers Initiative, where cognitive performance was assessed using five cognitive screening tools, including Symbol Digit Modalities Test (SDMT), Montreal Cognitive Assessment, Benton Judgment of Line Orientation, Modified Semantic Fluency Test, and Letter Number Sequencing Test, at baseline and subsequent annual follow-up visit for 5 years. Genetic data including ApoE and other PD risk genetic information were also obtained. We used SPSS-receiver operating characteristic and ANOVA repeated measures to evaluate which cognitive assessment is the best reflecting cognitive performance in PD at early stage and over time. Logistic regression analyses were used to determine the genetic associations with the rapidity of cognitive decline in PD. Results SDMT performed better in detecting mild cognitive impairment at baseline (AUC = 0.763), and SDMT was the only tool showing a steady cognitive decline during longitudinal observation. Multigenetic factors significantly associated with cognitive impairment at early stage of the disease (AUC = 0.950) with IP6K2 rs12497850 more evident, and a significantly faster decline (AUC = 0.831) within 5 years after motor onset, particularly in those carrying FGF20 rs591323. Conclusion SDMT is a preferable cognitive assessment tool for PD and genetic factors synergistically contribute to the cognitive dysfunction in PD.
Collapse
Affiliation(s)
- Ling-Xiao Cao
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wee Lee Kong
- Pharmacology Department, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Piu Chan
- Department of Neurobiology, Neurology and Geriatrics, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wei Zhang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Margaret J. Morris
- Pharmacology Department, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Yue Huang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Pharmacology Department, School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
5
|
Plzáková V, Mana J, Růžička E, Nikolai T. Efficacy of non-computerized cognitive rehabilitation in Parkinson's disease: A one year follow up study. APPLIED NEUROPSYCHOLOGY. ADULT 2024:1-12. [PMID: 38710156 DOI: 10.1080/23279095.2024.2341808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
In this study, we explored the effect of non-computerized cognitive rehabilitation in patients with Parkinson's disease in comparison with an intervention with elements of music therapy after the completion of a three-month program and one year after the end of the intervention. After the initial neuropsychological examination, the respondents were divided into two intervention groups. The experimental group (n = 26) underwent a twelve-week program of cognitive rehabilitation at a frequency of 60 minutes once a week. The control group (n = 27) underwent an intervention program with elements of music therapy at the same frequency. Respondents who underwent the cognitive rehabilitation program improved in the delayed recall from visual memory in the follow-up examination after the end of the cognitive intervention. One year after the end, the effect of cognitive rehabilitation persisted in delayed recall from visual memory and in executive mental flexibility. Cognitive rehabilitation is an effective approach to compensate for cognitive deficits in P D, but other approaches to cognitive stimulation may be equally effective.
Collapse
Affiliation(s)
- Vladimíra Plzáková
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine and General University Hospital in Prague, Charles University, Prague, Czech Republic
- Institute of Neuropsychiatric Care, Prague, Czech Republic
- Rehabilitation Hospital Beroun, Beroun, Czech Republic
| | - Josef Mana
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine and General University Hospital in Prague, Charles University, Prague, Czech Republic
| | - Evžen Růžička
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine and General University Hospital in Prague, Charles University, Prague, Czech Republic
| | - Tomáš Nikolai
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine and General University Hospital in Prague, Charles University, Prague, Czech Republic
- Department of Psychology, Faculty of Arts, Charles University, Prague, Czech Republic
| |
Collapse
|
6
|
Leitner C, D'Este G, Verga L, Rahayel S, Mombelli S, Sforza M, Casoni F, Zucconi M, Ferini-Strambi L, Galbiati A. Neuropsychological Changes in Isolated REM Sleep Behavior Disorder: A Systematic Review and Meta-analysis of Cross-sectional and Longitudinal Studies. Neuropsychol Rev 2024; 34:41-66. [PMID: 36588140 DOI: 10.1007/s11065-022-09572-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/28/2022] [Indexed: 01/03/2023]
Abstract
The aim of this meta-analysis is twofold: (a) to assess cognitive impairments in isolated rapid eye movement (REM) sleep behavior disorder (iRBD) patients compared to healthy controls (HC); (b) to quantitatively estimate the risk of developing a neurodegenerative disease in iRBD patients according to baseline cognitive assessment. To address the first aim, cross-sectional studies including polysomnography-confirmed iRBD patients, HC, and reporting neuropsychological testing were included. To address the second aim, longitudinal studies including polysomnography-confirmed iRBD patients, reporting baseline neuropsychological testing for converted and still isolated patients separately were included. The literature search was conducted based on PRISMA guidelines and the protocol was registered at PROSPERO (CRD42021253427). Cross-sectional and longitudinal studies were searched from PubMed, Web of Science, Scopus, and Embase databases. Publication bias and statistical heterogeneity were assessed respectively by funnel plot asymmetry and using I2. Finally, a random-effect model was performed to pool the included studies. 75 cross-sectional (2,398 HC and 2,460 iRBD patients) and 11 longitudinal (495 iRBD patients) studies were selected. Cross-sectional studies showed that iRBD patients performed significantly worse in cognitive screening scores (random-effects (RE) model = -0.69), memory (RE model = -0.64), and executive function (RE model = -0.50) domains compared to HC. The survival analyses conducted for longitudinal studies revealed that lower executive function and language performance, as well as the presence of mild cognitive impairment (MCI), at baseline were associated with an increased risk of conversion at follow-up. Our study underlines the importance of a comprehensive neuropsychological assessment in the context of iRBD.
Collapse
Affiliation(s)
- Caterina Leitner
- "Vita-Salute" San Raffaele University, Milan, Italy
- Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Via Stamira d'Ancona, 20, 20127, Milan, Italy
| | - Giada D'Este
- "Vita-Salute" San Raffaele University, Milan, Italy
- Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Via Stamira d'Ancona, 20, 20127, Milan, Italy
| | - Laura Verga
- Comparative Bioacoustics Group, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
- Faculty of Psychology and Neuroscience, Department NP&PP, Maastricht University, Maastricht, The Netherlands
| | - Shady Rahayel
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal, QC, Canada
- Center for Advanced Research in Sleep Medicine, Centre Intégré Universitaire de Santé et de Services Sociaux du Nord-de-l'Île-de-Montréal - Hôpital du Sacré-Cœur de Montréal, Montréal, QC, Canada
| | - Samantha Mombelli
- Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Via Stamira d'Ancona, 20, 20127, Milan, Italy
| | - Marco Sforza
- "Vita-Salute" San Raffaele University, Milan, Italy
- Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Via Stamira d'Ancona, 20, 20127, Milan, Italy
| | - Francesca Casoni
- Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Via Stamira d'Ancona, 20, 20127, Milan, Italy
| | - Marco Zucconi
- Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Via Stamira d'Ancona, 20, 20127, Milan, Italy
| | - Luigi Ferini-Strambi
- "Vita-Salute" San Raffaele University, Milan, Italy
- Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Via Stamira d'Ancona, 20, 20127, Milan, Italy
| | - Andrea Galbiati
- "Vita-Salute" San Raffaele University, Milan, Italy.
- Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Via Stamira d'Ancona, 20, 20127, Milan, Italy.
| |
Collapse
|
7
|
Ndayisaba A, Pitaro AT, Willett AS, Jones KA, de Gusmao CM, Olsen AL, Kim J, Rissanen E, Woods JK, Srinivasan SR, Nagy A, Nagy A, Mesidor M, Cicero S, Patel V, Oakley DH, Tuncali I, Taglieri-Noble K, Clark EC, Paulson J, Krolewski RC, Ho GP, Hung AY, Wills AM, Hayes MT, Macmore JP, Warren L, Bower PG, Langer CB, Kellerman LR, Humphreys CW, Glanz BI, Dielubanza EJ, Frosch MP, Freeman RL, Gibbons CH, Stefanova N, Chitnis T, Weiner HL, Scherzer CR, Scholz SW, Vuzman D, Cox LM, Wenning G, Schmahmann JD, Gupta AS, Novak P, Young GS, Feany MB, Singhal T, Khurana V. Clinical Trial-Ready Patient Cohorts for Multiple System Atrophy: Coupling Biospecimen and iPSC Banking to Longitudinal Deep-Phenotyping. CEREBELLUM (LONDON, ENGLAND) 2024; 23:31-51. [PMID: 36190676 PMCID: PMC9527378 DOI: 10.1007/s12311-022-01471-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/26/2022] [Indexed: 11/30/2022]
Abstract
Multiple system atrophy (MSA) is a fatal neurodegenerative disease of unknown etiology characterized by widespread aggregation of the protein alpha-synuclein in neurons and glia. Its orphan status, biological relationship to Parkinson's disease (PD), and rapid progression have sparked interest in drug development. One significant obstacle to therapeutics is disease heterogeneity. Here, we share our process of developing a clinical trial-ready cohort of MSA patients (69 patients in 2 years) within an outpatient clinical setting, and recruiting 20 of these patients into a longitudinal "n-of-few" clinical trial paradigm. First, we deeply phenotype our patients with clinical scales (UMSARS, BARS, MoCA, NMSS, and UPSIT) and tests designed to establish early differential diagnosis (including volumetric MRI, FDG-PET, MIBG scan, polysomnography, genetic testing, autonomic function tests, skin biopsy) or disease activity (PBR06-TSPO). Second, we longitudinally collect biospecimens (blood, CSF, stool) and clinical, biometric, and imaging data to generate antecedent disease-progression scores. Third, in our Mass General Brigham SCiN study (stem cells in neurodegeneration), we generate induced pluripotent stem cell (iPSC) models from our patients, matched to biospecimens, including postmortem brain. We present 38 iPSC lines derived from MSA patients and relevant disease controls (spinocerebellar ataxia and PD, including alpha-synuclein triplication cases), 22 matched to whole-genome sequenced postmortem brain. iPSC models may facilitate matching patients to appropriate therapies, particularly in heterogeneous diseases for which patient-specific biology may elude animal models. We anticipate that deeply phenotyped and genotyped patient cohorts matched to cellular models will increase the likelihood of success in clinical trials for MSA.
Collapse
Affiliation(s)
- Alain Ndayisaba
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Ariana T Pitaro
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Andrew S Willett
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Kristie A Jones
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Claudio Melo de Gusmao
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Abby L Olsen
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jisoo Kim
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Eero Rissanen
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jared K Woods
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Sharan R Srinivasan
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI , 48103, USA
| | - Anna Nagy
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Amanda Nagy
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Merlyne Mesidor
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Steven Cicero
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Viharkumar Patel
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Derek H Oakley
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Idil Tuncali
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Katherine Taglieri-Noble
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Emily C Clark
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jordan Paulson
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Richard C Krolewski
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Gary P Ho
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Albert Y Hung
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Anne-Marie Wills
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Michael T Hayes
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jason P Macmore
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | | | - Pamela G Bower
- The Multiple System Atrophy Coalition, Inc., 7918 Jones Branch Drive, Suite 300, McLean, VA, 22102, USA
| | - Carol B Langer
- The Multiple System Atrophy Coalition, Inc., 7918 Jones Branch Drive, Suite 300, McLean, VA, 22102, USA
| | - Lawrence R Kellerman
- The Multiple System Atrophy Coalition, Inc., 7918 Jones Branch Drive, Suite 300, McLean, VA, 22102, USA
| | - Christopher W Humphreys
- Department of Pulmonary, Sleep and Critical Care Medicine, Salem Hospital, MassGeneral Brigham, Salem, MA, 01970, USA
| | - Bonnie I Glanz
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Elodi J Dielubanza
- Department of Urology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Matthew P Frosch
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Roy L Freeman
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02115, USA
| | - Christopher H Gibbons
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02115, USA
| | - Nadia Stefanova
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Tanuja Chitnis
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Howard L Weiner
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Clemens R Scherzer
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Sonja W Scholz
- Laboratory of Neurogenetics, Disorders and Stroke, National Institute of Neurological, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, 21287, USA
| | - Dana Vuzman
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Laura M Cox
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Gregor Wenning
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Jeremy D Schmahmann
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Anoopum S Gupta
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Peter Novak
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Geoffrey S Young
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Tarun Singhal
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Vikram Khurana
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA.
| |
Collapse
|
8
|
Silva-Rodríguez J, Labrador-Espinosa MA, Moscoso A, Schöll M, Mir P, Grothe MJ. Characteristics of amnestic patients with hypometabolism patterns suggestive of Lewy body pathology. Brain 2023; 146:4520-4531. [PMID: 37284793 PMCID: PMC10629761 DOI: 10.1093/brain/awad194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/27/2023] [Accepted: 05/16/2023] [Indexed: 06/08/2023] Open
Abstract
A clinical diagnosis of Alzheimer's disease dementia (ADD) encompasses considerable pathological and clinical heterogeneity. While Alzheimer's disease patients typically show a characteristic temporo-parietal pattern of glucose hypometabolism on 18F-fluorodeoxyglucose (FDG)-PET imaging, previous studies have identified a subset of patients showing a distinct posterior-occipital hypometabolism pattern associated with Lewy body pathology. Here, we aimed to improve the understanding of the clinical relevance of these posterior-occipital FDG-PET patterns in patients with Alzheimer's disease-like amnestic presentations. Our study included 1214 patients with clinical diagnoses of ADD (n = 305) or amnestic mild cognitive impairment (aMCI, n = 909) from the Alzheimer's Disease Neuroimaging Initiative, who had FDG-PET scans available. Individual FDG-PET scans were classified as being suggestive of Alzheimer's (AD-like) or Lewy body (LB-like) pathology by using a logistic regression classifier trained on a separate set of patients with autopsy-confirmed Alzheimer's disease or Lewy body pathology. AD- and LB-like subgroups were compared on amyloid-β and tau-PET, domain-specific cognitive profiles (memory versus executive function performance), as well as the presence of hallucinations and their evolution over follow-up (≈6 years for aMCI, ≈3 years for ADD). Around 12% of the aMCI and ADD patients were classified as LB-like. For both aMCI and ADD patients, the LB-like group showed significantly lower regional tau-PET burden than the AD-like subgroup, but amyloid-β load was only significantly lower in the aMCI LB-like subgroup. LB- and AD-like subgroups did not significantly differ in global cognition (aMCI: d = 0.15, P = 0.16; ADD: d = 0.02, P = 0.90), but LB-like patients exhibited a more dysexecutive cognitive profile relative to the memory deficit (aMCI: d = 0.35, P = 0.01; ADD: d = 0.85 P < 0.001), and had a significantly higher risk of developing hallucinations over follow-up [aMCI: hazard ratio = 1.8, 95% confidence interval = (1.29, 3.04), P = 0.02; ADD: hazard ratio = 2.2, 95% confidence interval = (1.53, 4.06) P = 0.01]. In summary, a sizeable group of clinically diagnosed ADD and aMCI patients exhibit posterior-occipital FDG-PET patterns typically associated with Lewy body pathology, and these also show less abnormal Alzheimer's disease biomarkers as well as specific clinical features typically associated with dementia with Lewy bodies.
Collapse
Affiliation(s)
- Jesús Silva-Rodríguez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Miguel A Labrador-Espinosa
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - Alexis Moscoso
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Michael Schöll
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, 41345 Gothenburg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, WC1ELondon, UK
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, 41345 Gothenburg, Sweden
| | | |
Collapse
|
9
|
Bayram E, Batzu L, Tilley B, Gandhi R, Jagota P, Biundo R, Garon M, Prasertpan T, Lazcano-Ocampo C, Chaudhuri KR, Weil RS. Clinical trials for cognition in Parkinson's disease: Where are we and how can we do better? Parkinsonism Relat Disord 2023; 112:105385. [PMID: 37031010 PMCID: PMC10330317 DOI: 10.1016/j.parkreldis.2023.105385] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 03/24/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND Cognitive impairment is common in Parkinson's disease (PD) and has a substantial impact on quality of life. Despite numerous trials targeting various PD features, we still lack effective treatments for cognition beyond cholinesterase inhibitors. OBJECTIVE To identify the gaps in recent clinical trials with cognitive outcomes in PD and consider areas for improvement. METHODS We examined recent clinical trials with cognitive outcomes in PD registered on ClinicalTrials.gov, excluding trials without cognitive outcomes, non-interventional studies, and in atypical Parkinsonian disorders. Included trials were categorized by treatment approach (investigational medicinal product, behavioral, physical activity, device-based). Details of trial design and outcomes were collected. RESULTS 178 trials at different stages of trial completion were considered. 46 trials were completed, 25 had available results. Mean follow-up duration was 29.9 weeks. Most common cognitive measure was Montreal Cognitive Assessment. Most were performed in North America or Europe. Majority of the participants identified as non-Hispanic and White. Only eight trials showed improvement in cognition, none showed improvement beyond four months. These included trials of international medicinal products, cognitive and physical interventions and devices. GRADE certainty levels ranged from Moderate to Very Low. Only mevidalen had a Moderate certainty for potential clinical effectiveness. CONCLUSIONS Amongst a large number of trials for cognition in PD, only a small proportion were completed. Few showed significant improvement, with no proven long-lasting effects. Trial design, lack of enrichment for at-risk groups, short follow-up duration, insensitive outcome measures likely contribute to lack of detectable benefit and should be considered in future trials.
Collapse
Affiliation(s)
- Ece Bayram
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
| | - Lucia Batzu
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK.
| | - Bension Tilley
- Dementia Research Centre, University College London, London, UK; Department of Brain Sciences, Imperial College London, London, UK
| | - Rhea Gandhi
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Priya Jagota
- Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Roberta Biundo
- Department of General Psychology, University of Padua, Padua, Italy; Study Center for Neurodegeneration (CESNE), University of Padua, Padua, Italy
| | - Michela Garon
- Parkinson and Movement Disorders Unit, Department of Neuroscience, University of Padua, Padua, Italy
| | - Tittaya Prasertpan
- Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Claudia Lazcano-Ocampo
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Neurology, Hospital Sotero del Rio, Santiago, Chile
| | - K Ray Chaudhuri
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK
| | - Rimona S Weil
- Dementia Research Centre, University College London, London, UK; Movement Disorders Centre, University College London, London, UK
| |
Collapse
|
10
|
Schröter N, Bormann T, Rijntjes M, Blazhenets G, Berti R, Sajonz BE, Urbach H, Weiller C, Meyer PT, Rau A, Frings L. Cognitive Deficits in Parkinson's Disease Are Associated with Neuronal Dysfunction and Not White Matter Lesions. Mov Disord Clin Pract 2023; 10:1066-1073. [PMID: 37476309 PMCID: PMC10354622 DOI: 10.1002/mdc3.13792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 04/25/2023] [Accepted: 05/04/2023] [Indexed: 07/22/2023] Open
Abstract
Background Cognitive deficits considerably contribute to the patient's burden in Parkinson's disease (PD). While cognitive decline is linked to neuronal dysfunction, the additional role of white matter lesions (WML) is discussed controversially. Objective To investigate the influence of WML, in comparison to neuronal dysfunction, on cognitive deficits in PD. Methods We prospectively recruited patients with PD who underwent neuropsychological assessment using the Mattis Dementia Rating Scale 2 (DRS-2) or Parkinson Neuropsychometric Dementia Assessment (PANDA) and both MRI and PET with [18F]fluorodeoxyglucose (FDG). WML-load and PD cognition-related covariance pattern (PDCP) as a measure of neuronal dysfunction were read out. Relationship between cognitive performance and rank-transformed WML was analyzed with linear regression, controlling for the patients' age. PDCP subject scores were investigated likewise and in a second step adjusting for age and WML load. Results Inclusion criteria were met by 76 patients with a mean (± SD) age of 63.5 ± 9.0 years and disease duration of 10.7 ± 5.4 years. Neuropsychological testing revealed front executive and parietal deficits and a median DRS-2 score of 137 (range 119-144)/144 and PANDA score of 22 (range 3-30)/30. No association between WML and cognition was observed, whereas PDCP subject scores showed a trend-level negative correlation with the DRS-2 (P = 0.060) as well as a negative correlation with PANDA (P = 0.049) which persisted also after additional correction for WML (P = 0.039). Conclusion The present study indicates that microangiopathic WML do not have a relevant impact on neurocognitive performance in PD whereas neuronal dysfunction does.
Collapse
Affiliation(s)
- Nils Schröter
- Department of Neurology and Clinical Neuroscience, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Tobias Bormann
- Department of Neurology and Clinical Neuroscience, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Michel Rijntjes
- Department of Neurology and Clinical Neuroscience, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Ganna Blazhenets
- Department of Nuclear Medicine, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Raissa Berti
- Department of Nuclear Medicine, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Bastian E.A. Sajonz
- Department of Stereotactic and Functional Neurosurgery, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Horst Urbach
- Department of Neuroradiology, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Cornelius Weiller
- Department of Neurology and Clinical Neuroscience, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Philipp T. Meyer
- Department of Nuclear Medicine, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Alexander Rau
- Department of Neuroradiology, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Lars Frings
- Department of Nuclear Medicine, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Center for Geriatrics and Gerontology Freiburg, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| |
Collapse
|
11
|
McDermott AT, Frazier RL, Stewart PV. The Test of Visuospatial Construction: A Novel Test of Non-motoric Visuoconstruction. Arch Clin Neuropsychol 2022; 38:586-597. [DOI: 10.1093/arclin/acac087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2022] [Indexed: 11/18/2022] Open
Abstract
Abstract
Objective
The Test of Visuospatial Construction (TVSC) was designed as an easily administered measure of non-motor visuoconstruction, though only preliminary data exists regarding the clinical utility of this task. The current study examined the diagnostic accuracy of the TVSC by comparing performance between healthy subjects and various clinical groups. The authors also wanted to determine whether previous findings could be replicated regarding its effectiveness at tracking cognitive decline.
Method
Archival data collected over a period of more than 10 years were utilized and the overall sample consisted of 955 individuals, 372 healthy subjects, and 583 subjects who were categorized into various clinical groups. Only TVSC test data and demographic variables were utilized for statistical analyses in this study.
Results
The control group obtained significantly higher scores on the TVSC than the clinical groups. AUC values were indicative of excellent discrimination between cases and controls. Exploratory ROC curve analyses suggested adequate to excellent discrimination between the control group and the individual clinical groups as well as between the mild cognitive impairment (MCI) subgroups and the two dementia groups.
Conclusions
This study demonstrates that the TVSC can effectively differentiate between healthy subjects and neurologically compromised individuals. Additionally, the TVSC may be able to measure the progressive decline in visuoconstructive abilities that occurs as patients traverse the spectrum of MCI and dementia.
Collapse
Affiliation(s)
- Adam T McDermott
- Atrium Health Wake Forest Baptist Neuropsychology at Westchester, , High Point, NC , USA
| | - Rebecca L Frazier
- Atrium Health Wake Forest Baptist Neuropsychology at Westchester, , High Point, NC , USA
| | - Peter V Stewart
- Atrium Health Wake Forest Baptist Neuropsychology at Westchester, , High Point, NC , USA
| |
Collapse
|
12
|
Implication of EEG theta/alpha and theta/beta ratio in Alzheimer's and Lewy body disease. Sci Rep 2022; 12:18706. [PMID: 36333386 PMCID: PMC9636216 DOI: 10.1038/s41598-022-21951-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
Abstract
We evaluated the patterns of quantitative electroencephalography (EEG) in patients with Alzheimer's disease (AD), Lewy body disease (LBD), and mixed disease. Sixteen patients with AD, 38 with LBD, 20 with mixed disease, and 17 control participants were recruited and underwent EEG. The theta/alpha ratio and theta/beta ratio were measured. The relationship of the log-transformed theta/alpha ratio (TAR) and theta/beta ratio (TBR) with the disease group, the presence of AD and LBD, and clinical symptoms were evaluated. Participants in the LBD and mixed disease groups had higher TBR in all lobes except for occipital lobe than those in the control group. The presence of LBD was independently associated with higher TBR in all lobes and higher central and parietal TAR, while the presence of AD was not. Among cognitively impaired patients, higher TAR was associated with the language, memory, and visuospatial dysfunction, while higher TBR was associated with the memory and frontal/executive dysfunction. Increased TBR in all lobar regions and temporal TAR were associated with the hallucinations, while cognitive fluctuations and the severity of Parkinsonism were not. Increased TBR could be a biomarker for LBD, independent of AD, while the presence of mixed disease could be reflected as increased TAR.
Collapse
|
13
|
Yamada Y, Kobayashi M, Shinkawa K, Nemoto M, Ota M, Nemoto K, Arai T. Characteristics of Drawing Process Differentiate Alzheimer’s Disease and Dementia with Lewy Bodies. J Alzheimers Dis 2022; 90:693-704. [DOI: 10.3233/jad-220546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Early differential diagnosis of Alzheimer’s disease (AD) and dementia with Lewy bodies (DLB) is important for treatment and disease management, but it remains challenging. Although computer-based drawing analysis may help differentiate AD and DLB, it has not been extensively studied. Objective: We aimed to identify the differences in features characterizing the drawing process between AD, DLB, and cognitively normal (CN) individuals, and to evaluate the validity of using these features to identify and differentiate AD and DLB. Methods: We collected drawing data with a digitizing tablet and pen from 123 community-dwelling older adults in three clinical diagnostic groups of mild cognitive impairment or dementia due to AD (n = 47) or Lewy body disease (LBD; n = 27), and CN (n = 49), matched for their age, sex, and years of education. We then investigated drawing features in terms of the drawing speed, pressure, and pauses. Results: Reduced speed and reduced smoothness in speed and pressure were observed particularly in the LBD group, while increased pauses and total durations were observed in both the AD and LBD groups. Machine-learning models using these features achieved an area under the receiver operating characteristic curve (AUC) of 0.80 for AD versus CN, 0.88 for LBD versus CN, and 0.77 for AD versus LBD. Conclusion: Our results indicate how different types of drawing features were particularly discriminative between the diagnostic groups, and how the combination of these features can facilitate the identification and differentiation of AD and DLB.
Collapse
Affiliation(s)
| | | | | | - Miyuki Nemoto
- Department of Psychiatry, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Miho Ota
- Department of Psychiatry, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kiyotaka Nemoto
- Department of Psychiatry, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tetsuaki Arai
- Department of Psychiatry, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
14
|
Macoir J. The Cognitive and Language Profile of Dementia with Lewy Bodies. Am J Alzheimers Dis Other Demen 2022; 37:15333175221106901. [PMCID: PMC10581135 DOI: 10.1177/15333175221106901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Dementia with Lewy bodies (DLB) is a major neurocognitive disorder (MNCD) that is primarily characterized by motor, cognitive, and behavioral symptoms. Although not dominant in the clinical portrait of DLB, impairments affecting language processing have been reported. It is sometimes challenging to differentiate DLB from Alzheimer’s disease and dementia associated with Parkinson’s disease in clinical practice. Therefore, a better comprehension of the typical clinical presentation of DLB may be useful to ease the medical diagnosis. In this article, current data on cognitive and language disorders in DLB are reported, with special attention paid to their primary or secondary functional origin. The main elements that should be considered for the neuropsychological and speech-language assessment of individuals with possible or proven DLB are also highlighted. Additional studies are needed, especially for language impairment, to obtain a more accurate portrait of the clinical presentation of DLB and characterize its progression.
Collapse
Affiliation(s)
- Joël Macoir
- Département de Réadaptation, Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre de Recherche CERVO (CERVO Brain Research Centre), Québec, QC, Canada
| |
Collapse
|
15
|
Zhu Y, Yang B, Zhou C, Gao C, Hu Y, Yin WF, Yin K, Zhu Y, Jiang G, Ren H, Pang A, Yang X. Cortical atrophy is associated with cognitive impairment in Parkinson's disease: a combined analysis of cortical thickness and functional connectivity. Brain Imaging Behav 2022; 16:2586-2600. [PMID: 36044168 DOI: 10.1007/s11682-022-00714-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/28/2022] [Accepted: 08/07/2022] [Indexed: 11/30/2022]
Abstract
We aimed to perform a combined analysis of cortical thickness and functional connectivity to explore their association with cognitive impairment in Parkinson's disease (PD). A total of 53 PD and 15 healthy control subjects were enrolled. PD patients were divided into PD with normal cognition (PD-NC, n = 25), PD with mild cognitive impairment (PD-MCI, n = 11), and PD with dementia (PDD, n = 17). In some analyses, the PD-MCI and PDD groups were aggregated to represent "PD patients with cognitive impairment". Cognitive status was assessed with the Mini-Mental State Examination (MMSE). Anatomical magnetic resonance imaging and resting-state functional connectivity analysis were performed in all subjects. First, surface-based morphometry measurements of cortical thickness and voxels with cortical thickness reduction were detected. Then, regions showing reduced thickness were analyzed for changes in resting-state functional connectivity in PD involving cognitive impairment. Our results showed that, compared with PD-NC, patients with cognitive impairment showed decreased cortical thickness in the left superior temporal, left lingual, right insula, and right fusiform regions. PD-MCI patients showed these alterations in the right lingual region. Widespread cortical thinning was detected in PDD subjects, including the left superior temporal, left fusiform, right insula, and right fusiform areas. We found that cortical thinning in the left superior temporal, left fusiform, and right temporal pole regions positively correlated with MMSE score. In the resting-state functional connectivity analysis, we found a decrease in functional connectivity between the cortical atrophic brain areas mentioned above and cognition-related brain networks, as well as an increase in functional connectivity between those region and the cerebellum. Alterations in cortical thickness may result in a dysfunction of resting-state functional connectivity, contributing to cognitive decline in patients with PD. However, it is more probable that the relation between structure and FC would be bidirectional,and needs more research to explore in PD cognitve decline.
Collapse
Affiliation(s)
- Yongyun Zhu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, Yunnan Province, P.R. China
| | - Baiyuan Yang
- Department of Neurology, Seventh People's Hospital of Chengdu, 690041, Chengdu, Sichuan Province, P.R. China
| | - Chuanbin Zhou
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, Yunnan Province, P.R. China
| | - Chao Gao
- Department of medical imaging, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, Yunnan Province, P.R. China
| | - Yanfei Hu
- Department of medical imaging, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, Yunnan Province, P.R. China
| | - Wei Fang Yin
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, Yunnan Province, P.R. China
| | - Kangfu Yin
- Department of Neurology, Qujing City First People's Hospital, 655099, Qujing, Yunnan Province, P.R. China
| | - Yangfan Zhu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, Yunnan Province, P.R. China
| | - Guoliang Jiang
- Department of neurosurgery, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, Yunnan Province, P.R. China
| | - Hui Ren
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, Yunnan Province, P.R. China
| | - Ailan Pang
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, Yunnan Province, P.R. China.
| | - Xinglong Yang
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, Yunnan Province, P.R. China. .,Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, 650032, Kunming, Yunnan Province, P.R. China.
| |
Collapse
|
16
|
Hampsey E, Meszaros M, Skirrow C, Strawbridge R, Taylor RH, Chok L, Aarsland D, Al-Chalabi A, Chaudhuri R, Weston J, Fristed E, Podlewska A, Awogbemila O, Young AH. Protocol for Rhapsody: a longitudinal observational study examining the feasibility of speech phenotyping for remote assessment of neurodegenerative and psychiatric disorders. BMJ Open 2022; 12:e061193. [PMID: 35667724 PMCID: PMC9171270 DOI: 10.1136/bmjopen-2022-061193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/17/2022] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Neurodegenerative and psychiatric disorders (NPDs) confer a huge health burden, which is set to increase as populations age. New, remotely delivered diagnostic assessments that can detect early stage NPDs by profiling speech could enable earlier intervention and fewer missed diagnoses. The feasibility of collecting speech data remotely in those with NPDs should be established. METHODS AND ANALYSIS The present study will assess the feasibility of obtaining speech data, collected remotely using a smartphone app, from individuals across three NPD cohorts: neurodegenerative cognitive diseases (n=50), other neurodegenerative diseases (n=50) and affective disorders (n=50), in addition to matched controls (n=75). Participants will complete audio-recorded speech tasks and both general and cohort-specific symptom scales. The battery of speech tasks will serve several purposes, such as measuring various elements of executive control (eg, attention and short-term memory), as well as measures of voice quality. Participants will then remotely self-administer speech tasks and follow-up symptom scales over a 4-week period. The primary objective is to assess the feasibility of remote collection of continuous narrative speech across a wide range of NPDs using self-administered speech tasks. Additionally, the study evaluates if acoustic and linguistic patterns can predict diagnostic group, as measured by the sensitivity, specificity, Cohen's kappa and area under the receiver operating characteristic curve of the binary classifiers distinguishing each diagnostic group from each other. Acoustic features analysed include mel-frequency cepstrum coefficients, formant frequencies, intensity and loudness, whereas text-based features such as number of words, noun and pronoun rate and idea density will also be used. ETHICS AND DISSEMINATION The study received ethical approval from the Health Research Authority and Health and Care Research Wales (REC reference: 21/PR/0070). Results will be disseminated through open access publication in academic journals, relevant conferences and other publicly accessible channels. Results will be made available to participants on request. TRIAL REGISTRATION NUMBER NCT04939818.
Collapse
Affiliation(s)
- Elliot Hampsey
- Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
| | | | | | - Rebecca Strawbridge
- Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
| | - Rosie H Taylor
- Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
| | | | - Dag Aarsland
- Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
| | - Ammar Al-Chalabi
- Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
| | - Ray Chaudhuri
- Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital NHS Foundation Trust, London, UK
| | | | | | - Aleksandra Podlewska
- Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital NHS Foundation Trust, London, UK
| | - Olabisi Awogbemila
- Parkinson's Foundation Centre of Excellence, King's College Hospital NHS Foundation Trust, London, UK
| | - Allan H Young
- Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
| |
Collapse
|
17
|
Goldman JG, Holden SK. Cognitive Syndromes Associated With Movement Disorders. Continuum (Minneap Minn) 2022; 28:726-749. [PMID: 35678400 DOI: 10.1212/con.0000000000001134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE OF REVIEW This article reviews the recognition and management of cognitive syndromes in movement disorders, including those with parkinsonism, chorea, ataxia, dystonia, and tremor. RECENT FINDINGS Cognitive and motor syndromes are often intertwined in neurologic disorders, including neurodegenerative diseases such as Parkinson disease, atypical parkinsonian syndromes, Huntington disease, and other movement disorders. Cognitive symptoms often affect attention, working memory, and executive and visuospatial functions preferentially, rather than language and memory, but heterogeneity can be seen in the various movement disorders. A distinct cognitive syndrome has been recognized in patients with cerebellar syndromes. Appropriate recognition and screening for cognitive changes in movement disorders may play a role in achieving accurate diagnoses and guiding patients and their families regarding progression and management decisions. SUMMARY In the comprehensive care of patients with movement disorders, recognition of cognitive syndromes is important. Pharmacologic treatments for the cognitive syndromes, including mild cognitive impairment and dementia, in these movement disorders lag behind the therapeutics available for motor symptoms, and more research is needed. Patient evaluation and management require a comprehensive team approach, often linking neurologists as well as neuropsychologists, psychologists, psychiatrists, social workers, and other professionals.
Collapse
|
18
|
The role of the autonomic nervous system in cerebral blood flow regulation in dementia: A review. Auton Neurosci 2022; 240:102985. [DOI: 10.1016/j.autneu.2022.102985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 02/28/2022] [Accepted: 04/28/2022] [Indexed: 11/19/2022]
|
19
|
The Attention Network Test in Parkinson and Lewy Body Disease: A Systematic Review. Cogn Behav Neurol 2022; 35:1-13. [PMID: 35239595 DOI: 10.1097/wnn.0000000000000292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/27/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND The Attention Network Test (ANT) is a well-established measure of efficiency for the alerting, orienting, and executive attentional networks. However, its novel application in Parkinson disease (PD) and Lewy body dementia (LBD) research more broadly has yet to be evaluated systematically. OBJECTIVE To compare and consolidate the outcomes of studies reporting use of the ANT in PD and LBD groups and to identify the methodological considerations for the conduct of such studies. METHOD We performed a systematic literature search for articles exploring attention in PD and LBD groups using the ANT. We excluded articles on the basis of irrelevant scope, non-English, and groups other than PD and LBD. Once the full text articles were identified, we extracted the data and assessed the studies' quality. RESULTS The final sample included 16 articles ranging from low to moderate quality. Behavioral findings suggested a general slowing of responses yet preserved accuracy from the PD group compared with controls. Overall, the evidence was inconclusive regarding the state of the alerting network in the PD and LBD groups, mostly supportive of an intact orienting network, and strongly suggestive of an impaired executive network. Differences in sample stratification, patient symptomatology, and dopaminergic medication levels were identified as influential factors in the attentional results across studies. CONCLUSION Although sparse, the existing evidence indicates that the ANT is a viable option for measuring attention in PD; it can also be harnessed to explore the impact of symptoms and medications on attentional networks in PD and LBD groups.
Collapse
|
20
|
Hattori T, Reynolds R, Wiggs E, Horovitz SG, Lungu C, Chen G, Yasuda E, Hallett M. Neural correlates of working memory and compensation at different stages of cognitive impairment in Parkinson’s disease. NEUROIMAGE: CLINICAL 2022; 35:103100. [PMID: 35780660 PMCID: PMC9421432 DOI: 10.1016/j.nicl.2022.103100] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/09/2022] [Accepted: 06/23/2022] [Indexed: 11/22/2022] Open
Abstract
PD patients have two types of compensatory mechanisms for working memory (WM). First, hyperactivation for different WM load tasks depending on cognitive status. PD-CogNL has hyperactivation for moderate and heavy working memory load tasks. PD-MCI and PDD have hyperactivation for control and light working memory load tasks. Second, bilateral recruitment of WM-related areas improves WM performance.
Working memory (WM) impairment is one of the most frequent cognitive deficits in Parkinson's disease (PD). However, it is not known how neural activity is altered and compensatory responses eventually fail during progression. We aimed to elucidate neural correlates of WM and compensatory mechanisms in PD. Eighteen cognitively normal PD patients (PD-CogNL), 16 with PD with mild cognitive impairment (PD-MCI), 11 with PD with dementia (PDD), and 17 healthy controls (HCs) were evaluated. Subjects performed an n-back task. Functional MRI data were analyzed by event-related analysis for correct responses. Brain activations were evaluated by comparing them to fixation cross or 0-back task, and correlated with n-back task performance. When compared to fixation cross, PD-CogNL patients had more activation in WM areas than HCs for both the 2- and 3-back tasks. PD-MCI and PDD patients had more activation in WM areas than HCs for the 0- and 1-back task. 2-back task performance was correlated with brain activations (vs. 0-back task) in the bilateral dorsolateral prefrontal cortex and frontal eye field (FEF) and left rostral prefrontal cortex, caudate nucleus, inferior/superior parietal lobule (IPL/SPL), and anterior insular cortex as well as anterior cingulate cortex. 3-back task performance was correlated with brain activations (vs. 0-back task) in the left FEF, right caudate nucleus, and bilateral IPL/SPL. Additional activations on top of the 0-back task, rather than fixation cross, are the neural correlates of WM. Our results suggest PD patients have two types of compensatory mechanisms: (1) Hyperactivation for different WM load tasks depending on their cognitive status. PD-CogNL have hyperactivation for moderate and heavy working memory load tasks while maintaining normal working memory performance. In contrast, PD-MCI and PDD have hyperactivation for control task and light working memory load task, leaving less neural resources to further activate for more demanding tasks and resulting in impaired working memory performance. (2) Bilateral recruitment of WM-related areas, in particular the DLPFC, FEF, IPL/SPL and caudate nucleus, to improve WM performance.
Collapse
|
21
|
Palermo G, Belli E, Tommasini L, Morganti R, Frosini D, Nicoletti V, Tognoni G, Siciliano G, Bonuccelli U, Baldacci F, Ceravolo R. Dissecting the Interplay Between Time of Dementia and Cognitive Profiles in Lewy Body Dementias. J Alzheimers Dis 2021; 84:757-766. [PMID: 34602466 DOI: 10.3233/jad-210006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD) are differentiated by the time of onset of cognitive and motor symptoms ('1-year rule'). We explored the neuropsychological continuum of DLB and PDD subjects with different timing of dementia onset. OBJECTIVE Our aim was to compare the neuropsychological profile of DLB and PDD patients with different timing of dementia onset. METHODS Neuropsychological findings at the diagnosis of dementia of 66 PDD and 42 DLB patients were retrospectively compared. Patients with PDD were divided into three tertile subgroups according to the time interval between the onset of parkinsonism and dementia (N = 24, 2-4 years; N = 17, 5-7 years; N = 25 ≥8 years, respectively). RESULTS DLB patients performed worse on the Stroop and semantic fluency tests than PDD, even in comparison to PD with early dementia onset. No significant differences among PDD subgroups were reported. CONCLUSION Executive and semantic language tests could differentiate DLB and PD patients with earlier development of dementia relative to parkinsonism.
Collapse
Affiliation(s)
- Giovanni Palermo
- Department of Clinical and Experimental Medicine, Unit of Neurology, University of Pisa, Pisa, Italy
| | - Elisabetta Belli
- Department of Clinical and Experimental Medicine, Unit of Neurology, University of Pisa, Pisa, Italy
| | - Luca Tommasini
- Department of Clinical and Experimental Medicine, Unit of Neurology, University of Pisa, Pisa, Italy
| | | | - Daniela Frosini
- Department of Clinical and Experimental Medicine, Unit of Neurology, University of Pisa, Pisa, Italy
| | - Valentina Nicoletti
- Department of Clinical and Experimental Medicine, Unit of Neurology, University of Pisa, Pisa, Italy
| | - Gloria Tognoni
- Department of Clinical and Experimental Medicine, Unit of Neurology, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, Unit of Neurology, University of Pisa, Pisa, Italy
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, Unit of Neurology, University of Pisa, Pisa, Italy
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, Unit of Neurology, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, Unit of Neurology, University of Pisa, Pisa, Italy
| |
Collapse
|
22
|
Koros C, Stefanis L, Scarmeas N. Parkinsonism and dementia. J Neurol Sci 2021; 433:120015. [PMID: 34642023 DOI: 10.1016/j.jns.2021.120015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/01/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022]
Abstract
The aim of the present review is to summarize literature data on dementia in parkinsonian disorders. Cognitive decline and the gradual development of dementia are considered to be key features in the majority of parkinsonian conditions. The burden of dementia in everyday life of parkinsonian patients and their caregivers is vast and can be even more challenging to handle than the motor component of the disease. Common pathogenetic mechanisms involve the aggregation and spreading of abnormal proteins like alpha-synuclein, tau or amyloid in cortical and subcortical regions with subsequent dysregulation of multiple neurotransmitter systems. The degree of cognitive deterioration in these disorders is variable and ranges from mild cognitive impairment to severe cognitive dysfunction. There is also variation in the number and type of affected cognitive domains which can involve either a single domain like executive or visuospatial function or multiple ones. Novel genetic, biological fluid or imaging biomarkers appear promising in facilitating the diagnosis and staging of dementia in parkinsonian conditions. A significant part of current research in Parkinson's disease and other parkinsonian syndromes is targeted towards the cognitive aspects of these disorders. Stabilization or amelioration of cognitive outcomes represents a primary endpoint in many ongoing clinical trials for novel disease modifying treatments in this field. This article is part of the Special Issue "Parkinsonism across the spectrum of movement disorders and beyond" edited by Joseph Jankovic, Daniel D. Truong and Matteo Bologna.
Collapse
Affiliation(s)
- Christos Koros
- 1st Department of Neurology, Aeginition University, Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Leonidas Stefanis
- 1st Department of Neurology, Aeginition University, Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece; Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aeginition University, Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece; The Gertrude H. Sergievsky Center, Department of Neurology, Taub Institute for Research in Alzheimer's, Disease and the Aging Brain, Columbia University, New York, USA.
| |
Collapse
|
23
|
What Do We Know about Theory of Mind Impairment in Parkinson's Disease? Behav Sci (Basel) 2021; 11:bs11100130. [PMID: 34677223 PMCID: PMC8533307 DOI: 10.3390/bs11100130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022] Open
Abstract
Theory of mind (ToM) is a social cognitive skill that involves the ability to attribute mental states to self and others (what they think (cognitive ToM) and feel (affective ToM)). We aim to provide an overview of previous knowledge of ToM in Parkinson's disease (PD). In the last few years more attention has been paid to the study of this construct as a non-motor manifestation of PD. In advanced stages, both components of ToM (cognitive and affective) are commonly impaired, although in early PD results remain controversial. Executive dysfunction correlates with ToM deficits and other cognitive domains such as language and visuospatial function have also been related to ToM. Recent studies have demonstrated that PD patients with mild cognitive impairment show ToM deficits more frequently in comparison with cognitively normal PD patients. In addition to the heterogeneity of ToM tests administered in different studies, depression and dopaminergic medication may also be acting as confounding factors, but there are still insufficient data to support this. Neuroimaging studies conducted to understand the underlying networks of cognitive and affective ToM deficits in PD are lacking. The study of ToM deficit in PD continues to be important, as this may worsen quality of life and favor social stigma. Future studies should be considered, including assessment of the patients' cognitive state, associated mood disorders, and the role of dopaminergic deficit.
Collapse
|
24
|
Writers Are Common among Parkinson's Disease Patients: A Longitudinal Study. Behav Neurol 2021; 2021:5559926. [PMID: 34035867 PMCID: PMC8121563 DOI: 10.1155/2021/5559926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/22/2021] [Accepted: 05/04/2021] [Indexed: 11/26/2022] Open
Abstract
Parkinson's disease (PD) patients may have a specific personality profile, which includes being introvert, cautious and devoted to hard work. The evaluation of psychological characteristics must be evaluated according to methods for assessments of personality disorders. Such evaluations are often time-consuming and available only in research settings. The “parkinsonian trait” may be established early in life but may change with disease progression. To overcome this long interval before onset of PD questions on literary activities were included in the medical record. Three percent of PD patients could be defined as writers, significantly higher than observed in the general population. PD writers published their first books long before onset of disease. Being a writer is an extrovert trait meaning that the patient is prepared for criticism and publicity. We suggest that questions regarding personal activities prior to disease onset add valuable information on personality which differs significantly from traits observed later in the disease period.
Collapse
|
25
|
Howard E, Irwin DJ, Rascovsky K, Nevler N, Shellikeri S, Tropea TF, Spindler M, Deik A, Chen-Plotkin A, Siderowf A, Dahodwala N, Weintraub D, Shaw LM, Trojanowski JQ, Vaishnavi SN, Wolk DA, Mechanic-Hamilton D, Morley JF, Duda JE, Grossman M, Cousins KAQ. Cognitive Profile and Markers of Alzheimer Disease-Type Pathology in Patients With Lewy Body Dementias. Neurology 2021; 96:e1855-e1864. [PMID: 33593865 PMCID: PMC8105963 DOI: 10.1212/wnl.0000000000011699] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To determine whether patients with Lewy body dementia (LBD) with likely Alzheimer disease (AD)-type copathology are more impaired on confrontation naming than those without likely AD-type copathology. METHODS We selected 57 patients with LBD (dementia with Lewy bodies [DLB], n = 38; Parkinson disease dementia [PDD], n = 19) with available AD CSF biomarkers and neuropsychological data. CSF β-amyloid1-42 (Aβ42), phosphorylated-tau (p-tau), and total-tau (t-tau) concentrations were measured. We used an autopsy-validated CSF cut point (t-tau:Aβ42 ratio > 0.3, n = 43), or autopsy data when available (n = 14), to categorize patients as having LBD with (LBD + AD, n = 26) and without (LBD - AD, n = 31) likely AD-type copathology. Analysis of covariance tested between-group comparisons across biologically defined groups (LBD + AD, LBD - AD) and clinical phenotypes (DLB, PDD) on confrontation naming (30-item Boston Naming Test [BNT]), executive abilities (letter fluency [LF], reverse digit span [RDS]), and global cognition (Mini-Mental State Examination [MMSE]), with adjustment for age at dementia onset, time from dementia onset to test date, and time from CSF to test date. Spearman correlation related cognitive performance to CSF analytes. RESULTS Patients with LBD + AD performed worse on BNT than patients with LBD - AD (F = 4.80, p = 0.03); both groups performed similarly on LF, RDS, and MMSE (all p > 0.1). Clinically defined PDD and DLB groups did not differ in performance on any of these measures (all p > 0.05). A correlation across all patients showed that BNT score was negatively associated with CSF t-tau (ρ = -0.28, p < 0.05) and p-tau (ρ = -0.26, p = 0.05) but not Aβ42 (p > 0.1). CONCLUSION Markers of AD-type copathology are implicated in impaired language performance in LBD. Biologically based classification of LBD may be advantageous over clinically defined syndromes to elucidate clinical heterogeneity.
Collapse
Affiliation(s)
- Erica Howard
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - David J Irwin
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Katya Rascovsky
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Naomi Nevler
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Sanjana Shellikeri
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Thomas F Tropea
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Meredith Spindler
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Andres Deik
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Alice Chen-Plotkin
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Andrew Siderowf
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Nabila Dahodwala
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Daniel Weintraub
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Leslie M Shaw
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - John Q Trojanowski
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Sanjeev N Vaishnavi
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - David A Wolk
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Dawn Mechanic-Hamilton
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - James F Morley
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - John E Duda
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Murray Grossman
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA
| | - Katheryn A Q Cousins
- From the Department of Neurology (E.H., D.J.I., K.R., N.N., S.S., T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W., S.N.V., D.A.W., D.M.-H., J.F.M., J.E.D., M.G., K.A.Q.C.), Frontotemporal Degeneration Center (E.H., D.J.I., K.R., N.N., S.S., M.G., K.A.Q.C.), Parkinson's Disease and Movement Disorders Center (T.F.T., M.S., A.D., A.C.-P., A.S., N.D., D.W.), Digital Neuropathology Laboratory (D.J.I.), Alzheimer's Disease Center (J.Q.T., S.N.V., D.A.W., D.M.-H.), Center for Neurodegenerative Disease Research (L.M.S., J.Q.T.), and Department of Pathology and Laboratory Medicine (L.M.S., J.Q.T., D.A.W.), Perelman School of Medicine at the University of Pennsylvania; and Michael J. Crescenz VA Medical Center (D.W., J.F.M., J.E.D.), Parkinson's Disease Research, Education, and Clinical Center, Philadelphia, PA.
| |
Collapse
|
26
|
Cong C, Zhang W, Qian X, Qiu W, Ma C. Significant Overlap of α-Synuclein, Amyloid-β, and Phospho-Tau Pathologies in Neuropathological Diagnosis of Lewy-related Pathology: Evidence from China Human Brain Bank. J Alzheimers Dis 2021; 80:447-458. [PMID: 33554920 DOI: 10.3233/jad-201548] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Lewy-related pathology (LRP), primarily comprised of α-synuclein, is a typical neuropathological change that has been identified in many neurodegenerative disorders such as Parkinson's disease (PD), PD with dementia, and dementia with Lewy bodies. OBJECTIVE To investigate the distribution of LRP in the China Human Brain Bank, the co-occurrence of neuropathologic features of Alzheimer's disease (AD) in LRP cases, and LRP-related cognitive dysfunction. METHODS LRP neuropathological diagnosis was performed in 180 postmortem brains. AD neuropathological diagnosis was then performed in the 21 neuropathologically-diagnosed LRP cases. Antemortem cognitive functioning evaluation (Everyday Cognitive, ECog) was assessed for brain donors by the immediate kin of the donor within 24 hours after death. RESULTS 12% (21 in 180) postmortem brains were neuropathologically diagnosed as LRP cases. 86% (18 in 21) aged above 80, 81% (17 in 21) LRP cases combined with AD neuropathology, and 62% (13 in 21) combined with both the intermediate or high-level amyloid-β and phospho-tau pathologies. ECog scores showed significant differences between the groups of LRP brainstem-predominant type and LRP diffuse neocortical type, and between groups of AD and the combined LRP (diffuse neocortical type)-AD. CONCLUSION The overlap of neocortical α-synuclein, amyloid-β, phospho-tau, and neuritic plaques in LRP suggested the potential interplay among the common characteristics of proteinopathies in the late stage of neuropathological development of LRP in human brains. The anatomic progression of LRP, the process of α-synuclein spreading from the brainstem to limbic and neocortical regions, might aggravate the deterioration of cognitive function in addition to that effect of AD.
Collapse
Affiliation(s)
- Cong Cong
- Institute of Basic Medical Sciences, Neuroscience Center, National Human Brain Bank for Development and Function, Chinese Academy of Medical Sciences, Beijing, China.,Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wanying Zhang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,Department of Respiratory Medicine, Capital Medical University, Beijing, China
| | - Xiaojing Qian
- Institute of Basic Medical Sciences, Neuroscience Center, National Human Brain Bank for Development and Function, Chinese Academy of Medical Sciences, Beijing, China.,Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wenying Qiu
- Institute of Basic Medical Sciences, Neuroscience Center, National Human Brain Bank for Development and Function, Chinese Academy of Medical Sciences, Beijing, China.,Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Chao Ma
- Institute of Basic Medical Sciences, Neuroscience Center, National Human Brain Bank for Development and Function, Chinese Academy of Medical Sciences, Beijing, China.,Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College, Beijing, China.,Chinese Institute for Brain Research, Beijing, China
| |
Collapse
|
27
|
Rouse HJ, Small BJ, Schinka JA, Loewenstein DA, Duara R, Potter H. Mild behavioral impairment as a predictor of cognitive functioning in older adults. Int Psychogeriatr 2021; 33:285-293. [PMID: 32456733 DOI: 10.1017/s1041610220000678] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To assess the influence of mild behavioral impairment (MBI) on the cognitive performance of older adults who are cognitively healthy or have mild cognitive impairment (MCI). METHODS Secondary data analysis of a sample (n = 497) of older adults from the Florida Alzheimer's Disease Research Center who were either cognitively healthy (n = 285) or diagnosed with MCI (n = 212). Over half of the sample (n = 255) met the operationalized diagnostic criteria for MBI. Cognitive domains of executive function, attention, short-term memory, and episodic memory were assessed using a battery of neuropsychological tests. RESULTS Older adults with MBI performed worse on tasks of executive function, attention, and episodic memory compared to those without MBI. A significant interaction revealed that persons with MBI and MCI performed worse on tasks of episodic memory compared to individuals with only MCI, but no significant differences were found in performance in cognitively healthy older adults with or without MBI on this cognitive domain. As expected, cognitively healthy older adults performed better than individuals with MCI on every domain of cognition. CONCLUSIONS The present study found evidence that independent of cognitive status, individuals with MBI performed worse on tests of executive function, attention, and episodic memory than individuals without MBI. Additionally, those with MCI and MBI perform significantly worse on episodic memory tasks than individuals with only MCI. These results provide support for a unique cognitive phenotype associated with MBI and highlight the necessity for assessing both cognitive and behavioral symptoms.
Collapse
Affiliation(s)
- Hillary J Rouse
- School of Aging Studies, University of South Florida, Tampa, FL, USA
| | - Brent J Small
- School of Aging Studies, University of South Florida, Tampa, FL, USA
| | - John A Schinka
- School of Aging Studies, University of South Florida, Tampa, FL, USA
| | - David A Loewenstein
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, University of Miami, Miami, FL, USA
| | - Ranjan Duara
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, University of Miami, Miami, FL, USA
| | - Huntington Potter
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
28
|
Ramezani M, Mouches P, Yoon E, Rajashekar D, Ruskey JA, Leveille E, Martens K, Kibreab M, Hammer T, Kathol I, Maarouf N, Sarna J, Martino D, Pfeffer G, Gan-Or Z, Forkert ND, Monchi O. Investigating the relationship between the SNCA gene and cognitive abilities in idiopathic Parkinson's disease using machine learning. Sci Rep 2021; 11:4917. [PMID: 33649398 PMCID: PMC7921412 DOI: 10.1038/s41598-021-84316-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/10/2021] [Indexed: 01/16/2023] Open
Abstract
Cognitive impairments are prevalent in Parkinson's disease (PD), but the underlying mechanisms of their development are unknown. In this study, we aimed to predict global cognition (GC) in PD with machine learning (ML) using structural neuroimaging, genetics and clinical and demographic characteristics. As a post-hoc analysis, we aimed to explore the connection between novel selected features and GC more precisely and to investigate whether this relationship is specific to GC or is driven by specific cognitive domains. 101 idiopathic PD patients had a cognitive assessment, structural MRI and blood draw. ML was performed on 102 input features including demographics, cortical thickness and subcortical measures, and several genetic variants (APOE, MAPT, SNCA, etc.). Using the combination of RRELIEFF and Support Vector Regression, 11 features were found to be predictive of GC including sex, rs894280, Edinburgh Handedness Inventory, UPDRS-III, education, five cortical thickness measures (R-parahippocampal, L-entorhinal, R-rostral anterior cingulate, L-middle temporal, and R-transverse temporal), and R-caudate volume. The rs894280 of SNCA gene was selected as the most novel finding of ML. Post-hoc analysis revealed a robust association between rs894280 and GC, attention, and visuospatial abilities. This variant indicates a potential role for the SNCA gene in cognitive impairments of idiopathic PD.
Collapse
Affiliation(s)
- Mehrafarin Ramezani
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Pauline Mouches
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Eunjin Yoon
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Deepthi Rajashekar
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Jennifer A Ruskey
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Etienne Leveille
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Kristina Martens
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mekale Kibreab
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Tracy Hammer
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Iris Kathol
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nadia Maarouf
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Justyna Sarna
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Davide Martino
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Gerald Pfeffer
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ziv Gan-Or
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Nils D Forkert
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Oury Monchi
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Hotchkiss Brain Institute (HBI), Cummings School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Radiology, University of Calgary, Calgary, AB, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
| |
Collapse
|
29
|
Ruppert MC, Greuel A, Freigang J, Tahmasian M, Maier F, Hammes J, van Eimeren T, Timmermann L, Tittgemeyer M, Drzezga A, Eggers C. The default mode network and cognition in Parkinson's disease: A multimodal resting-state network approach. Hum Brain Mapp 2021; 42:2623-2641. [PMID: 33638213 PMCID: PMC8090788 DOI: 10.1002/hbm.25393] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
Involvement of the default mode network (DMN) in cognitive symptoms of Parkinson's disease (PD) has been reported by resting-state functional MRI (rsfMRI) studies. However, the relation to metabolic measures obtained by [18F]-fluorodeoxyglucose positron emission tomography (FDG-PET) is largely unknown. We applied multimodal resting-state network analysis to clarify the association between intrinsic metabolic and functional connectivity abnormalities within the DMN and their significance for cognitive symptoms in PD. PD patients were classified into normal cognition (n = 36) and mild cognitive impairment (MCI; n = 12). The DMN was identified by applying an independent component analysis to FDG-PET and rsfMRI data of a matched subset (16 controls and 16 PD patients) of the total cohort. Besides metabolic activity, metabolic and functional connectivity within the DMN were compared between the patients' groups and healthy controls (n = 16). Glucose metabolism was significantly reduced in all DMN nodes in both patient groups compared to controls, with the lowest uptake in PD-MCI (p < .05). Increased metabolic and functional connectivity along fronto-parietal connections was identified in PD-MCI patients compared to controls and unimpaired patients. Functional connectivity negatively correlated with cognitive composite z-scores in patients (r = -.43, p = .005). The current study clarifies the commonalities of metabolic and hemodynamic measures of brain network activity and their individual significance for cognitive symptoms in PD, highlighting the added value of multimodal resting-state network approaches for identifying prospective biomarkers.
Collapse
Affiliation(s)
- Marina C Ruppert
- Department of Neurology, University Hospital of Marburg, Marburg, Germany.,Center for Mind, Brain, and Behavior-CMBB, Universities of Marburg and Gießen, Marburg, Germany
| | - Andrea Greuel
- Department of Neurology, University Hospital of Marburg, Marburg, Germany
| | - Julia Freigang
- Department of Neurology, University Hospital of Marburg, Marburg, Germany.,Center for Mind, Brain, and Behavior-CMBB, Universities of Marburg and Gießen, Marburg, Germany
| | - Masoud Tahmasian
- Institute of Medical Science and Technology, Shahid Beheshti University, Tehran, Iran
| | - Franziska Maier
- Medical Faculty, Department of Psychiatry, University Hospital Cologne, Cologne, Germany
| | - Jochen Hammes
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Cologne, Germany
| | - Thilo van Eimeren
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Cologne, Germany.,Department of Neurology, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Cologne, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lars Timmermann
- Department of Neurology, University Hospital of Marburg, Marburg, Germany.,Center for Mind, Brain, and Behavior-CMBB, Universities of Marburg and Gießen, Marburg, Germany
| | - Marc Tittgemeyer
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Cluster of Excellence in Cellular Stress and Aging Associated Disease (CECAD), Cologne, Germany
| | - Alexander Drzezga
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Cologne, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-2), Jülich, Germany
| | - Carsten Eggers
- Department of Neurology, University Hospital of Marburg, Marburg, Germany.,Center for Mind, Brain, and Behavior-CMBB, Universities of Marburg and Gießen, Marburg, Germany
| |
Collapse
|
30
|
Khedr EM, Mohamed KO, Ali AM, Hasan AM. The effect of repetitive transcranial magnetic stimulation on cognitive impairment in Parkinson's disease with dementia: Pilot study. Restor Neurol Neurosci 2021; 38:55-66. [PMID: 31815705 DOI: 10.3233/rnn-190956] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The exact mechanism of cognitive impairment in PD is not known. Repetitive transcranial magnetic stimulation (rTMS) has been proposed as a possible treatment for cognitive impairment and to treat the motor symptoms in Parkinson's disease (PD) where its effects seem additive to those of dopaminergic medications. OBJECTIVE In this pilot study we investigated whether repeated sessions of rTMS have an effect on measures of cognitive impairment in patients with PD dementia. METHODS 33 patients with PD dementia were randomly assigned sham or real rTMS (2000 pulses; 20 Hz; 90% RMT; 10 trains of 10 s with 25 s between each train) over the hand area of each motor cortex (5 min between hemispheres) for 10 days (5 days/week) followed by 5 booster sessions every month for 3 months. Assessments included the Unified Parkinson's Disease Rating Scale part III (UPDRS), Montreal Cognitive Assessment (MoCA); Mini Mental State Examination (MMSE), Clinical Dementia Rating Scale (CDR); Memory and Executive Screening (MES) and Instrumental activity of Daily Living (IADL). Event related potentials (P300) and cortical excitability were measured before treatment and after the last session. RESULTS There were no significant differences in the effects of rTMS between groups. Although rTMS improved motor function in the active group it had only a minor effect on two of the dementia rating scores (the MMSE and MoCA) but not the others (CDR and MES). There was also a reduction in the latency of the P300 in the active group. CONCLUSIONS rTMS over M1 is useful for motor function and may have a small positive effect on cognition. However, better approaches for the latter are necessary, may be require multisite rTMS to target both motor and frontal cortical region.
Collapse
Affiliation(s)
- Eman M Khedr
- Department of Neuropsychiatry, Assiut University Hospital, Assiut, Egypt
| | - Khaled O Mohamed
- Department of Neuropsychiatry, Assiut University Hospital, Assiut, Egypt
| | - Anwar M Ali
- Department of Neuropsychiatry, Assiut University Hospital, Assiut, Egypt
| | - Asmaa M Hasan
- Department of Neuropsychiatry, Assiut University Hospital, Assiut, Egypt
| |
Collapse
|
31
|
Orad RI, Shiner T. Differentiating dementia with Lewy bodies from Alzheimer's disease and Parkinson's disease dementia: an update on imaging modalities. J Neurol 2021; 269:639-653. [PMID: 33511432 DOI: 10.1007/s00415-021-10402-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 12/16/2022]
Abstract
Dementia with Lewy bodies is the second most common cause of neurodegenerative dementia after Alzheimer's disease. Dementia with Lewy bodies can provide a diagnostic challenge due to the frequent overlap of clinical signs with other neurodegenerative conditions, namely Parkinson's disease dementia, and Alzheimer's disease. Part of this clinical overlap is due to the neuropathological overlap. Dementia with Lewy bodies is characterized by the accumulation of aggregated α-synuclein protein in Lewy bodies, similar to Parkinson's disease and Parkinson's disease dementia. However, it is also frequently accompanied by aggregation of amyloid-beta and tau, the pathological hallmarks of Alzheimer's disease. Neuroimaging is central to the diagnostic process. This review is an overview of both established and evolving imaging methods that can improve diagnostic accuracy and improve management of this disorder.
Collapse
Affiliation(s)
- Rotem Iris Orad
- Cognitive Neurology Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, 6, Weismann St, Tel Aviv, Israel. .,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Tamara Shiner
- Cognitive Neurology Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, 6, Weismann St, Tel Aviv, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Movement Disorders Unit, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
32
|
Wilson H, de Natale ER, Politis M. Nucleus basalis of Meynert degeneration predicts cognitive impairment in Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:189-205. [DOI: 10.1016/b978-0-12-819975-6.00010-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
|
33
|
Armstrong MJ, Irwin DJ, Leverenz JB, Gamez N, Taylor A, Galvin JE. Biomarker Use for Dementia With Lewy Body Diagnosis: Survey of US Experts. Alzheimer Dis Assoc Disord 2021; 35:55-61. [PMID: 33009039 PMCID: PMC7904569 DOI: 10.1097/wad.0000000000000414] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/08/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Dementia with Lewy body (DLB) diagnostic criteria define "indicative" and "supportive" biomarkers, but clinical practice patterns are unknown. METHODS An anonymous survey querying clinical use of diagnostic tests/biomarkers was sent to 38 center of excellence investigators. The survey included "indicative" biomarkers (dopamine transporter scan, myocardial scintigraphy, polysomnography), "supportive" biomarkers [magnetic resonance imaging (MRI)], positron emission tomography, or single-photon emission computed tomography perfusion/metabolism scans, quantitative electroencephalography), and other diagnostic tests (neuropsychological testing, cerebrospinal fluid analysis, genetics). Responses were analyzed descriptively. RESULTS Of the 22 respondents (58%), all reported the capability to perform neuropsychological testing, MRI, polysomnography, dopamine transporter scans, positron emission tomography/single-photon emission computed tomography scans, and cerebrospinal fluid analysis; 96% could order genetic testing. Neuropsychological testing and MRI were the most commonly ordered tests. Diagnostic testing beyond MRI and neuropsychological testing was most helpful in the context of "possible" DLB and mild cognitive impairment and to assist with differential diagnosis. Myocardial scintigraphy and electroencephalograpy use were rare. CONCLUSIONS AND RELEVANCE Neuropsychological testing and MRI remain the most widely used diagnostic tests by DLB specialists. Other tests-particularly indicative biomarkers-are used only selectively. Research is needed to validate existing potential DLB biomarkers, develop new biomarkers, and investigate mechanisms to improve DLB diagnosis.
Collapse
Affiliation(s)
- Melissa J. Armstrong
- Department of Neurology, University of Florida College of Medicine, McKnight Brain Institute
| | | | - James B. Leverenz
- Cleveland Lou Ruvo Center for Brain Health – Neurological Institute, Cleveland Clinic
| | - Noheli Gamez
- Department of Neurology, University of Florida College of Medicine, McKnight Brain Institute
| | | | - James E. Galvin
- Comprehensive Center for Brain Health, Department of Neurology, University of Miami Miller School of Medicine
| |
Collapse
|
34
|
Mood and emotional disorders associated with parkinsonism, Huntington disease, and other movement disorders. HANDBOOK OF CLINICAL NEUROLOGY 2021; 183:175-196. [PMID: 34389117 DOI: 10.1016/b978-0-12-822290-4.00015-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This chapter provides a review of mood, emotional disorders, and emotion processing deficits associated with diseases that cause movement disorders, including Parkinson's disease, Lewy body dementia, multiple system atrophy, progressive supranuclear palsy, corticobasal degeneration, frontotemporal dementia with parkinsonism, Huntington's disease, essential tremor, dystonia, and tardive dyskinesia. For each disorder, a clinical description of the common signs and symptoms, disease progression, and epidemiology is provided. Then the mood and emotional disorders associated with each of these diseases are described and discussed in terms of clinical presentation, incidence, prevalence, and alterations in quality of life. Alterations of emotion communication, such as affective speech prosody and facial emotional expression, associated with these disorders are also discussed. In addition, if applicable, deficits in gestural and lexical/verbal emotion are reviewed. Throughout the chapter, the relationships among mood and emotional disorders, alterations of emotional experiences, social communication, and quality of life, as well as treatment, are emphasized.
Collapse
|
35
|
Resting state activity and connectivity of the nucleus basalis of Meynert and globus pallidus in Lewy body dementia and Parkinson's disease dementia. Neuroimage 2020; 221:117184. [PMID: 32711059 PMCID: PMC7762815 DOI: 10.1016/j.neuroimage.2020.117184] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/19/2020] [Accepted: 07/16/2020] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB) are two related diseases which can be difficult to distinguish. There is no objective biomarker which can reliably differentiate between them. The synergistic combination of electrophysiological and neuroimaging approaches is a powerful method for interrogation of functional brain networks in vivo. We recorded bilateral local field potentials (LFPs) from the nucleus basalis of Meynert (NBM) and the internal globus pallidus (GPi) with simultaneous cortical magnetoencephalography (MEG) in six PDD and five DLB patients undergoing surgery for deep brain stimulation (DBS) to look for differences in underlying resting-state network pathophysiology. In both patient groups we observed spectral peaks in the theta (2–8 Hz) band in both the NBM and the GPi. Furthermore, both the NBM and the GPi exhibited similar spatial and spectral patterns of coupling with the cortex in the two disease states. Specifically, we report two distinct coherent networks between the NBM/GPi and cortical regions: (1) a theta band (2–8 Hz) network linking the NBM/GPi to temporal cortical regions, and (2) a beta band (13–22 Hz) network coupling the NBM/GPi to sensorimotor areas. We also found differences between the two disease groups: oscillatory power in the low beta (13–22Hz) band was significantly higher in the globus pallidus in PDD patients compared to DLB, and coherence in the high beta (22–35Hz) band between the globus pallidus and lateral sensorimotor cortex was significantly higher in DLB patients compared to PDD. Overall, our findings reveal coherent networks of the NBM/GPi region that are common to both DLB and PDD. Although the neurophysiological differences between the two conditions in this study are confounded by systematic differences in DBS lead trajectories and motor symptom severity, they lend support to the hypothesis that DLB and PDD, though closely related, are distinguishable from a neurophysiological perspective.
Collapse
|
36
|
Pourzinal D, Yang JHJ, Byrne GJ, O'Sullivan JD, Mitchell L, McMahon KL, Copland DA, Dissanayaka NN. Identifying subtypes of mild cognitive impairment in Parkinson's disease using cluster analysis. J Neurol 2020; 267:3213-3222. [PMID: 32535681 DOI: 10.1007/s00415-020-09977-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/01/2020] [Accepted: 06/04/2020] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The concept of Mild Cognitive Impairment (MCI) in Parkinson's disease (PD) has shown the potential for identifying at-risk dementia patients. Identifying subtypes of MCI is likely to assist therapeutic discoveries and better clinical management of patients with PD (PWP). Recent cluster-based approaches have demonstrated dominance in memory and executive impairment in PD. The present study will further explore the role of memory and executive impairment and associated clinical features in non-demented PWP. METHOD A K-means cluster analysis was performed on ten "frontal" and "posterior" cognitive variables derived from a dataset of 85 non-demented PWP. The resulting cluster structure was chosen based on quantitative, qualitative, theoretical, and clinical validity. Cluster profiles were then created through statistical analysis of cognitive and clinical/demographic variables. A descriptive analysis of each cluster's performance on a comprehensive PD-MCI diagnostic battery was also explored. RESULTS The resulting cluster structure revealed four distinct cognitive phenotypes: (1) frontal-dominant impairment; (2) posterior-cortical-dominant impairment; (3) global impairment, and (4) cognitively intact. Demographic profiling revealed significant differences in the age, gender split, global cognitive ability, and motor symptoms between these clusters. However, there were no significant differences between the clusters on measures of depression, apathy, and anxiety. CONCLUSION These results validate the existence of distinct cognitive phenotypes within PD-MCI and encourage future research into their clinical trajectory and neuroimaging correlates.
Collapse
Affiliation(s)
- Dana Pourzinal
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital, Building 71/918, Herston, Brisbane, QLD, 4029, Australia
- School of Psychology, The University of Queensland, St Lucia, Brisbane, QLD, 4067, Australia
| | - Ji Hyun J Yang
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital, Building 71/918, Herston, Brisbane, QLD, 4029, Australia
| | - Gerard J Byrne
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital, Building 71/918, Herston, Brisbane, QLD, 4029, Australia
- Mental Health Service, Royal Brisbane and Women's Hospital, Herston, Brisbane, QLD, 4029, Australia
| | - John D O'Sullivan
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital, Building 71/918, Herston, Brisbane, QLD, 4029, Australia
- Department of Neurology, Royal Brisbane and Women's Hospital, Herston, Brisbane, QLD, 4029, Australia
| | - Leander Mitchell
- School of Psychology, The University of Queensland, St Lucia, Brisbane, QLD, 4067, Australia
| | - Katie L McMahon
- School of Clinical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, 4000, Australia
| | - David A Copland
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital, Building 71/918, Herston, Brisbane, QLD, 4029, Australia
- School of Health and Rehabilitation Sciences, The University of Queensland, St Lucia, Brisbane, QLD, 4067, Australia
| | - Nadeeka N Dissanayaka
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital, Building 71/918, Herston, Brisbane, QLD, 4029, Australia.
- School of Psychology, The University of Queensland, St Lucia, Brisbane, QLD, 4067, Australia.
- Department of Neurology, Royal Brisbane and Women's Hospital, Herston, Brisbane, QLD, 4029, Australia.
| |
Collapse
|
37
|
Boertien JM, van der Zee S, Chrysou A, Gerritsen MJJ, Jansonius NM, Spikman JM, van Laar T. Study protocol of the DUtch PARkinson Cohort (DUPARC): a prospective, observational study of de novo Parkinson's disease patients for the identification and validation of biomarkers for Parkinson's disease subtypes, progression and pathophysiology. BMC Neurol 2020; 20:245. [PMID: 32534583 PMCID: PMC7293131 DOI: 10.1186/s12883-020-01811-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
Background Parkinson’s Disease (PD) is a heterogeneous, progressive neurodegenerative disorder which is characterized by a variety of motor and non-motor symptoms. To date, no disease modifying treatment for PD exists. Here, the study protocol of the Dutch Parkinson Cohort (DUPARC) is described. DUPARC is a longitudinal cohort study aimed at deeply phenotyping de novo PD patients who are treatment-naïve at baseline, to discover and validate biomarkers for PD progression, subtypes and pathophysiology. Methods/design DUPARC is a prospective cohort study in which 150 de novo PD subjects will be recruited through a collaborative network of PD treating neurologists in the northern part of the Netherlands (Parkinson Platform Northern Netherlands, PPNN). Participants will receive follow-up assessments after 1 year and 3 years, with the intention of an extended follow-up with 3 year intervals. Subjects are extensively characterized to primarily assess objectives within three major domains of PD: cognition, gastrointestinal function and vision. This includes brain magnetic resonance imaging (MRI); brain cholinergic PET-imaging with fluoroethoxybenzovesamicol (FEOBV-PET); brain dopaminergic PET-imaging with fluorodopa (FDOPA-PET); detailed neuropsychological assessments, covering all cognitive domains; gut microbiome composition; intestinal wall permeability; optical coherence tomography (OCT); genotyping; motor and non-motor symptoms; overall clinical status and lifestyle factors, including a dietary assessment; storage of blood and feces for additional analyses of inflammation and metabolic parameters. Since the start of the inclusion, at the end of 2017, over 100 PD subjects with a confirmed dopaminergic deficit on FDOPA-PET have been included. Discussion DUPARC is the first study to combine data within, but not limited to, the non-motor domains of cognition, gastrointestinal function and vision in PD subjects over time. As a de novo PD cohort, with treatment naïve subjects at baseline, DUPARC provides a unique opportunity for biomarker discovery and validation without the possible confounding influences of dopaminergic medication. Trial registration NCT04180865; registered retrospectively, November 28th 2019.
Collapse
Affiliation(s)
- Jeffrey M Boertien
- Department of Neurology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands.,Parkinson Expertise Center Groningen, Groningen, the Netherlands
| | - Sygrid van der Zee
- Department of Neurology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands.,Parkinson Expertise Center Groningen, Groningen, the Netherlands
| | - Asterios Chrysou
- Department of Neurology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands.,Parkinson Expertise Center Groningen, Groningen, the Netherlands
| | - Marleen J J Gerritsen
- Department of Neuropsychology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Nomdo M Jansonius
- Department of Ophthalmology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jacoba M Spikman
- Department of Neuropsychology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Teus van Laar
- Department of Neurology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands. .,Parkinson Expertise Center Groningen, Groningen, the Netherlands.
| | | |
Collapse
|
38
|
Colloby SJ, Watson R, Blamire AM, O’Brien JT, Taylor JP. Cortical thinning in dementia with Lewy bodies and Parkinson disease dementia. Aust N Z J Psychiatry 2020; 54:633-643. [PMID: 31696728 PMCID: PMC7285984 DOI: 10.1177/0004867419885165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND We investigated the structural changes associated with Alzheimer's disease, dementia with Lewy bodies and Parkinson disease dementia by means of cortical thickness analysis. METHODS Two hundred and forty-five participants: 76 Alzheimer's disease, 65 dementia with Lewy bodies, 29 Parkinson disease dementia and 76 cognitively normal controls underwent 3-T T1-weighted magnetic resonance imaging and clinical and cognitive assessments. We implemented FreeSurfer to obtain cortical thickness estimates to contrast patterns of cortical thinning across groups and their clinical correlates. RESULTS In Alzheimer's disease and dementia with Lewy bodies, a largely similar pattern of regional cortical thinning was observed relative to controls apart from a more severe loss within the entorhinal and parahippocampal structures in Alzheimer's disease. In Parkinson disease dementia, regional cortical thickness was indistinguishable from controls and dementia with Lewy bodies, suggesting an 'intermediate' pattern of regional cortical change. In terms of global cortical thickness, group profiles were controls > Parkinson disease dementia > dementia with Lewy bodies > Alzheimer's disease (F3, 241 ⩽ 123.2, p < 0.001), where percentage wise, the average difference compared to controls were -1.8%, -5.5% and -6.4%, respectively. In these samples, cortical thinning was also associated with cognitive decline in dementia with Lewy bodies but not in Parkinson disease dementia and Alzheimer's disease. CONCLUSION In a large and well-characterised cohort of people with dementia, regional cortical thinning in dementia with Lewy bodies was broadly similar to Alzheimer's disease. There was preservation of the medial temporal lobe structures in dementia with Lewy bodies compared with Alzheimer's disease, supporting its inclusion as a supportive biomarker in the revised clinical criteria for dementia with Lewy bodies. However, there was less global cortical thinning in Parkinson disease dementia, with no significant regional difference between Parkinson disease dementia and controls. These findings highlight the overlap across the Alzheimer's disease/Parkinson disease dementia spectrum and the potential for differing mechanisms underlying neurodegeneration and cognition in dementia with Lewy bodies and Parkinson disease dementia.
Collapse
Affiliation(s)
- Sean J Colloby
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK,Sean J Colloby, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK.
| | - Rosie Watson
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia,Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew M Blamire
- Institute of Cellular Medicine and Newcastle Magnetic Resonance Centre, Newcastle University, Newcastle upon Tyne, UK
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - John-Paul Taylor
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
39
|
Gratwicke J, Zrinzo L, Kahan J, Peters A, Brechany U, McNichol A, Beigi M, Akram H, Hyam J, Oswal A, Day B, Mancini L, Thornton J, Yousry T, Crutch SJ, Taylor JP, McKeith I, Rochester L, Schott JM, Limousin P, Burn D, Rossor MN, Hariz M, Jahanshahi M, Foltynie T. Bilateral nucleus basalis of Meynert deep brain stimulation for dementia with Lewy bodies: A randomised clinical trial. Brain Stimul 2020; 13:1031-1039. [PMID: 32334074 DOI: 10.1016/j.brs.2020.04.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 03/02/2020] [Accepted: 04/15/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dementia with Lewy bodies (DLB) is the second most common form of dementia. Current symptomatic treatment with medications remains inadequate. Deep brain stimulation of the nucleus basalis of Meynert (NBM DBS) has been proposed as a potential new treatment option in dementias. OBJECTIVE To assess the safety and tolerability of low frequency (20 Hz) NBM DBS in DLB patients and explore its potential effects on both clinical symptoms and functional connectivity in underlying cognitive networks. METHODS We conducted an exploratory randomised, double-blind, crossover trial of NBM DBS in six DLB patients recruited from two UK neuroscience centres. Patients were aged between 50 and 80 years, had mild-moderate dementia symptoms and were living with a carer-informant. Patients underwent image guided stereotactic implantation of bilateral DBS electrodes with the deepest contacts positioned in the Ch4i subsector of NBM. Patients were subsequently assigned to receive either active or sham stimulation for six weeks, followed by a two week washout period, then the opposite condition for six weeks. Safety and tolerability of both the surgery and stimulation were systematically evaluated throughout. Exploratory outcomes included the difference in scores on standardised measurements of cognitive, psychiatric and motor symptoms between the active and sham stimulation conditions, as well as differences in functional connectivity in discrete cognitive networks on resting state fMRI. RESULTS Surgery and stimulation were well tolerated by all six patients (five male, mean age 71.33 years). One serious adverse event occurred: one patient developed antibiotic-associated colitis, prolonging his hospital stay by two weeks. No consistent improvements were observed in exploratory clinical outcome measures, but the severity of neuropsychiatric symptoms reduced with NBM DBS in 3/5 patients. Active stimulation was associated with functional connectivity changes in both the default mode network and the frontoparietal network. CONCLUSION Low frequency NBM DBS can be safely conducted in DLB patients. This should encourage further exploration of the possible effects of stimulation on neuropsychiatric symptoms and corresponding changes in functional connectivity in cognitive networks. TRIAL REGISTRATION NUMBER NCT02263937.
Collapse
Affiliation(s)
- James Gratwicke
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK.
| | - Ludvic Zrinzo
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Joshua Kahan
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Amy Peters
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Una Brechany
- Biomedical Research Building, Newcastle University & Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Ann McNichol
- Biomedical Research Building, Newcastle University & Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Mazda Beigi
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Harith Akram
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Jonathan Hyam
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Ashwini Oswal
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Brian Day
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Laura Mancini
- Lynsholm Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - John Thornton
- Lynsholm Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Tarek Yousry
- Lynsholm Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Sebastian J Crutch
- Dementia Research Centre, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - John-Paul Taylor
- Newcastle University & Northumberland, Tyne and Wear NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Ian McKeith
- Newcastle University & Northumberland, Tyne and Wear NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Lynn Rochester
- Biomedical Research Building, Newcastle University & Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Jonathan M Schott
- Dementia Research Centre, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Patricia Limousin
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - David Burn
- Biomedical Research Building, Newcastle University & Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Martin N Rossor
- Dementia Research Centre, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Marwan Hariz
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Marjan Jahanshahi
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Thomas Foltynie
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London, UK.
| |
Collapse
|
40
|
Del Pino R, Murueta-Goyena A, Acera M, Carmona-Abellan M, Tijero B, Lucas-Jiménez O, Ojeda N, Ibarretxe-Bilbao N, Peña J, Gabilondo I, Gómez-Esteban JC. Autonomic dysfunction is associated with neuropsychological impairment in Lewy body disease. J Neurol 2020; 267:1941-1951. [PMID: 32170444 DOI: 10.1007/s00415-020-09783-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE This study aimed to analyze the association of autonomic dysfunction with cognition, depression, apathy, and fatigue in Lewy body disease (LBD). METHODS We included 61 patients [49 with idiopathic Parkinson's disease, 7 with dementia with Lewy bodies, and 5 E46K-SNCA mutation carriers] and 22 healthy controls. All participants underwent a comprehensive battery of neuropsychological and clinical measures, autonomic symptom assessment with the SCOPA-AUT, analysis of non-invasive hemodynamic parameters during deep breathing, the Valsalva maneuver, and a 20-min tilt test, and electrochemical skin conductance measurement at rest (Sudoscan). Student's t tests were used to assess group differences, and bivariate correlations and stepwise linear regressions to explore associations between autonomic function, cognition, depression, apathy, and fatigue. RESULTS Compared to controls, patients who had significant impairment (p < 0.05) in cognition, higher depression, apathy, and fatigue, more autonomic symptoms and objective autonomic dysfunction, reduced deep breathing heart rate variability [expiratory-to-inspiratory (E/I) ratio], prolonged pressure recovery time, and lower blood pressure in Valsalva late phase II and phase IV, while 24.1% had orthostatic hypotension in the tilt test. Autonomic parameters significantly correlated with cognitive and neuropsychiatric outcomes, systolic blood pressure during the Valsalva maneuver predicting apathy and depression. The E/I ratio was the main predictor of cognitive performance (17.6% for verbal fluency to 32.8% for visual memory). CONCLUSION Cardiovascular autonomic dysfunction is associated with cognitive and neuropsychiatric impairment in LBD, heart rate variability during deep breathing and systolic blood pressure changes during the Valsalva procedure are the main predictors of neuropsychological performance and depression/apathy symptoms, respectively.
Collapse
Affiliation(s)
- Rocío Del Pino
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Plaza de Cruces 12, CP, 48903, Barakaldo, Bizkaia, Spain. .,International University of La Rioja, La Rioja, Spain.
| | - Ane Murueta-Goyena
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Plaza de Cruces 12, CP, 48903, Barakaldo, Bizkaia, Spain
| | - Marian Acera
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Plaza de Cruces 12, CP, 48903, Barakaldo, Bizkaia, Spain
| | - Mar Carmona-Abellan
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Plaza de Cruces 12, CP, 48903, Barakaldo, Bizkaia, Spain
| | - Beatriz Tijero
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Plaza de Cruces 12, CP, 48903, Barakaldo, Bizkaia, Spain.,Neurology Department, Cruces University Hospital, Barakaldo, Bizkaia, Spain
| | - Olaia Lucas-Jiménez
- Department of Methods and Experimental Psychology, Faculty of Psychology and Education, University of Deusto, Bilbao, Spain
| | - Natalia Ojeda
- Department of Methods and Experimental Psychology, Faculty of Psychology and Education, University of Deusto, Bilbao, Spain
| | - Naroa Ibarretxe-Bilbao
- Department of Methods and Experimental Psychology, Faculty of Psychology and Education, University of Deusto, Bilbao, Spain
| | - Javier Peña
- Department of Methods and Experimental Psychology, Faculty of Psychology and Education, University of Deusto, Bilbao, Spain
| | - Iñigo Gabilondo
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Plaza de Cruces 12, CP, 48903, Barakaldo, Bizkaia, Spain.,Neurology Department, Cruces University Hospital, Barakaldo, Bizkaia, Spain.,Ikerbasque: The Basque Foundation for Science, Bilbao, Spain
| | - Juan Carlos Gómez-Esteban
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Plaza de Cruces 12, CP, 48903, Barakaldo, Bizkaia, Spain.,Neurology Department, Cruces University Hospital, Barakaldo, Bizkaia, Spain.,Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
41
|
Abstract
OBJECTIVES Dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD) have substantial clinical and biological overlap, with cognitive deficits typically observed in the executive and visuospatial domains. However, the neuropsychological profiles of mild cognitive impairment (MCI) associated with these disorders are not well understood. METHODS This systematic review examined existing literature on cognition in MCI due to LB disease (MCI-LB) and PD (PD-MCI) using an electronic search of seven databases (Medline, Embase, Psychinfo, PubMed, ProQuest, Scopus, and ScienceDirect). MCI-LB results were reviewed narratively given the small number of resulting papers (n = 7). Outcome variables from PD-MCI studies (n = 13) were extracted for meta-analysis of standardised mean differences (SMD). RESULTS In MCI-LB, executive dysfunction and slowed processing speed were the most prominent impairments, while visuospatial and working memory (WM) functions were also poor. MCI-LB scored significantly lower on verbal memory tests relative to controls, but significantly higher than patients with MCI due to Alzheimer's disease. Quantitative analysis of studies in PD-MCI showed a similar profile of impairment, with the largest deficits in visuospatial function (Benton Judgement of Line Orientation, SMD g = -2.09), executive function (Trail Making Test B, SMD g = -1.65), verbal ability (Naming Tests, SMD g = -0.140), and WM (Trail Making Test A, SMD g = -1.20). In both MCI-LB and PD-MCI, verbal and visuospatial memory retrieval was impaired, while encoding and storage appeared relatively intact. CONCLUSIONS The findings of this systematic review indicate similar neuropsychological profiles in the MCI stages of DLB and PDD. Executive impairment may at least partially explain poor performance in other domains.
Collapse
|
42
|
Auclair-Ouellet N, Mandl S, Kibreab M, Haffenden A, Hanganu A, Cheetham J, Kathol I, Sarna J, Martino D, Monchi O. Characterization of cognition in mild cognitive impairment with and without Parkinson's disease. Clin Park Relat Disord 2020; 3:100034. [PMID: 34316620 PMCID: PMC8298772 DOI: 10.1016/j.prdoa.2020.100034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 11/24/2022] Open
Abstract
Screening tests can diagnose PD-MCI but do not give detailed cognitive profiles. Criteria based on a complete neuropsychological battery identify more PD patients with MCI. The overall cognitive profile is similar in PD-MCI and MCI. Neuropsychological batteries and definition of impairment cut-offs should be refined.
Collapse
Affiliation(s)
- N Auclair-Ouellet
- School of Communication Sciences and Disorders, Faculty of Medicine, McGill University, Montreal, Canada.,Centre for Research on Brain, Language and Music, Montreal, Canada.,Centre de recherche de l'Institut universitaire de gériatrie de Montréal, Montreal, Canada
| | - S Mandl
- Centre for Research on Brain, Language and Music, Montreal, Canada.,Centre de recherche de l'Institut universitaire de gériatrie de Montréal, Montreal, Canada.,Cognitive Science Program, Faculty of Arts and Faculty of Science, McGill University, Montreal, Canada
| | - M Kibreab
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - A Haffenden
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - A Hanganu
- Centre de recherche de l'Institut universitaire de gériatrie de Montréal, Montreal, Canada.,Département de Psychologie, Faculté des Arts et des Sciences, Université de Montréal, Montreal, Canada
| | - J Cheetham
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - I Kathol
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - J Sarna
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - D Martino
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - O Monchi
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Département de Radiologie, Faculté de Médecine, Université de Montréal, Montreal, Canada
| |
Collapse
|
43
|
Loniewska MM, Gupta A, Bhatia S, MacKay-Clackett I, Jia Z, Wells PG. DNA damage and synaptic and behavioural disorders in glucose-6-phosphate dehydrogenase-deficient mice. Redox Biol 2020; 28:101332. [PMID: 31581069 PMCID: PMC6812046 DOI: 10.1016/j.redox.2019.101332] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/11/2019] [Accepted: 09/17/2019] [Indexed: 12/27/2022] Open
Abstract
Mice deficient in glucose-6-phosphate dehydrogenase (G6PD) cannot replenish the cellular antioxidant glutathione, which detoxifies neurodegenerative reactive oxygen species (ROS). To determine the functional consequences of G6PD deficiency, young and aging G6PD-deficient mice were evaluated for brain G6PD activity, DNA damage (comets, γH2AX), Purkinje cell loss, brain function (electrophysiology, behaviour) and lifespan. DNA comet formation was increased and Purkinje cell counts were decreased in a G6pd gene dose-dependent fashion. γH2AX formation varied by age, sex and brain region, with increased levels in G6PD-deficient young and aging females, and in aging males. Aging male G6PD-deficient mice exhibited synaptic dysfunction in hippocampal slices. G6PD-deficient young and aging females exhibited deficits in executive function, and young deficient mice exhibited deficits in social dominance. Conversely, median lifespan in G6PD-deficient females and males was enhanced. Enhanced ROS-initiated brain damage in G6PD deficiency has functional consequences, suggesting that G6PD protects against ROS-mediated neurodegenerative disorders.
Collapse
Affiliation(s)
- Margaret M Loniewska
- Faculty of Pharmacy and Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Anmol Gupta
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Shama Bhatia
- Faculty of Pharmacy and Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Isabel MacKay-Clackett
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Zhengping Jia
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Peter G Wells
- Faculty of Pharmacy and Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
44
|
Bee Venom: Overview of Main Compounds and Bioactivities for Therapeutic Interests. Molecules 2019; 24:molecules24162997. [PMID: 31430861 PMCID: PMC6720840 DOI: 10.3390/molecules24162997] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/14/2019] [Accepted: 08/16/2019] [Indexed: 11/17/2022] Open
Abstract
Apitherapy is an alternate therapy that relies on the usage of honeybee products, most importantly bee venom for the treatment of many human diseases. The venom can be introduced into the human body by manual injection or by direct bee stings. Bee venom contains several active molecules such as peptides and enzymes that have advantageous potential in treating inflammation and central nervous system diseases, such as Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis. Moreover, bee venom has shown promising benefits against different types of cancer as well as anti-viral activity, even against the challenging human immunodeficiency virus (HIV). Many studies described biological activities of bee venom components and launched preclinical trials to improve the potential use of apitoxin and its constituents as the next generation of drugs. The aim of this review is to summarize the main compounds of bee venom, their primary biological properties, mechanisms of action, and their therapeutic values in alternative therapy strategies.
Collapse
|
45
|
Hansen D, Ling H, Lashley T, Holton JL, Warner TT. Review: Clinical, neuropathological and genetic features of Lewy body dementias. Neuropathol Appl Neurobiol 2019; 45:635-654. [PMID: 30977926 DOI: 10.1111/nan.12554] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 04/09/2019] [Indexed: 01/08/2023]
Abstract
Lewy body dementias are the second most common neurodegenerative dementias after Alzheimer's disease and include dementia with Lewy bodies and Parkinson's disease dementia. They share similar clinical and neuropathological features but differ in the time of dementia and parkinsonism onset. Although Lewy bodies are their main pathological hallmark, several studies have shown the emerging importance of Alzheimer's disease pathology. Clinical amyloid-β imaging using Pittsburgh Compound B (PiB) supports neuropathological studies which found that amyloid-β pathology is more common in dementia with Lewy bodies than in Parkinson's disease dementia. Nevertheless, other co-occurring pathologies, such as cerebral amyloid angiopathy, TDP-43 pathology and synaptic pathology may also influence the development of neurodegeneration and dementia. Recent genetic studies demonstrated an important role of APOE genotype and other genes such as GBA and SNCA which seem to be involved in the pathophysiology of Lewy body dementias. The aim of this article is to review the main clinical, neuropathological and genetic aspects of dementia with Lewy bodies and Parkinson's disease dementia. This is particularly relevant as future management for these two conditions may differ.
Collapse
Affiliation(s)
- D Hansen
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
| | - H Ling
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK.,Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - T Lashley
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - J L Holton
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - T T Warner
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK.,Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
46
|
Liu AKL, Chau TW, Lim EJ, Ahmed I, Chang RCC, Kalaitzakis ME, Graeber MB, Gentleman SM, Pearce RKB. Hippocampal CA2 Lewy pathology is associated with cholinergic degeneration in Parkinson's disease with cognitive decline. Acta Neuropathol Commun 2019; 7:61. [PMID: 31023342 PMCID: PMC6485180 DOI: 10.1186/s40478-019-0717-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 04/10/2019] [Indexed: 01/06/2023] Open
Abstract
Although the precise neuropathological substrates of cognitive decline in Parkinson's disease (PD) remain elusive, it has long been regarded that pathology in the CA2 hippocampal subfield is characteristic of Lewy body dementias, including dementia in PD (PDD). Early non-human primate tracer studies demonstrated connections from the nucleus of the vertical limb of the diagonal band of Broca (nvlDBB, Ch2) to the hippocampus. However, the relationship between Lewy pathology of the CA2 subfield and cholinergic fibres has not been explored. Therefore, in this study, we investigated the burden of pathology in the CA2 subsector of PD cases with varying degrees of cognitive impairment and correlated this with the extent of septohippocampal cholinergic deficit. Hippocampal sections from 67 PD, 34 PD with mild cognitive impairment and 96 PDD cases were immunostained for tau and alpha-synuclein, and the respective pathology burden was assessed semi-quantitatively. In a subset of cases, the degree of CA2 cholinergic depletion was quantified using confocal microscopy and correlated with cholinergic neuronal loss in Ch2. We found that only cases with dementia have a significantly greater Lewy pathology, whereas cholinergic fibre depletion was evident in cases with mild cognitive impairment and this was significantly correlated with loss of cholinergic neurons in Ch2. In addition, multiple antigen immunofluorescence demonstrated colocalisation between cholinergic fibres and alpha-synuclein but not tau pathology. Such specific Lewy pathology targeting the cholinergic system within the CA2 subfield may contribute to the unique memory retrieval deficit seen in patients with Lewy body disorders, as distinct from the memory storage deficit seen in Alzheimer's disease.
Collapse
Affiliation(s)
- Alan King Lun Liu
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK.
| | - Tsz Wing Chau
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Ernest Junwei Lim
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Idil Ahmed
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China
| | - Michail E Kalaitzakis
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Manuel B Graeber
- Brain and Mind Centre, Bosch Institute, Discipline of Anatomy and Embryology, and Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Steve M Gentleman
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Ronald K B Pearce
- Neuropathology Unit, Division of Brain Sciences, Department of Medicine, Imperial College London, 4/F, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
47
|
Sestini S, Alongi P, Berti V, Calcagni ML, Cecchin D, Chiaravalloti A, Chincarini A, Cistaro A, Guerra UP, Pappatà S, Tiraboschi P, Nobili F. The role of molecular imaging in the frame of the revised dementia with Lewy body criteria. Clin Transl Imaging 2019. [DOI: 10.1007/s40336-019-00321-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
48
|
Kang SW, Jeon S, Yoo HS, Chung SJ, Lee PH, Sohn YH, Yun M, Evans AC, Ye BS. Effects of Lewy body disease and Alzheimer disease on brain atrophy and cognitive dysfunction. Neurology 2019; 92:e2015-e2026. [PMID: 30944239 DOI: 10.1212/wnl.0000000000007373] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/07/2019] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES To investigate the independent and interaction effects of Alzheimer disease (AD) and Lewy body disease (LBD) on cognition and brain atrophy. METHODS We consecutively recruited 38 controls and 108 patients with AD-related cognitive impairment (ADCI) and/or LBD-related cognitive impairment (LBCI) from university-based dementia and movement clinics. Diagnoses of ADCI and LBCI were supported by 18F-florbetaben PET and 18F-N-(3-fluoropropyl)-2β-carbon ethoxy-3β-(4-iodophenyl) nortropane-PET, respectively. There were 38 controls, 26 patients with pure ADCI (18 mild cognitive impairment [MCI] and 8 dementia), 28 patients with pure LBCI (13 MCI and 15 dementia), and 54 patients with mixed ADCI and LBCI (17 MCI and 37 dementia). We performed group-wise comparisons for neuropsychological z scores and regional cortical thickness. We also evaluated the effects of ADCI and LBCI using general linear models. RESULTS Compared to the controls, patients in the pure ADCI group and pure LBCI group had focused cortical thinning in the bilateral entorhinal/right anterior temporal cortices and bilateral anteromedial temporal/basal frontal cortices, respectively, while the mixed disease group had additional cortical thinning in the widespread association cortices. The independent effects of ADCI and LBCI on regional cortical thinning overlapped in the widespread association cortices, especially at the bilateral temporoparietal junction and parietal cortices. ADCI and LBCI had independent detrimental effects on the copying item of the Rey-Osterrieth Complex Figure Test. CONCLUSIONS Concomitant ADCI and LBCI are associated with the accentuation of neurodegeneration to widespread association cortices, and both diseases contribute to visuospatial dysfunction.
Collapse
Affiliation(s)
- Sung Woo Kang
- From the Departments of Neurology (S.W.K., H.S.Y., S.J.C., P.H.L., Y.H.S., B.S.Y.) and Nuclear Medicine (M.Y.), Yonsei University College of Medicine, Seoul, Korea; and McGill Centre for Integrative Neuroscience (S.J., A.C.E.), Montreal Neurological Institute, McGill University, Quebec, Canada
| | - Seun Jeon
- From the Departments of Neurology (S.W.K., H.S.Y., S.J.C., P.H.L., Y.H.S., B.S.Y.) and Nuclear Medicine (M.Y.), Yonsei University College of Medicine, Seoul, Korea; and McGill Centre for Integrative Neuroscience (S.J., A.C.E.), Montreal Neurological Institute, McGill University, Quebec, Canada
| | - Han Soo Yoo
- From the Departments of Neurology (S.W.K., H.S.Y., S.J.C., P.H.L., Y.H.S., B.S.Y.) and Nuclear Medicine (M.Y.), Yonsei University College of Medicine, Seoul, Korea; and McGill Centre for Integrative Neuroscience (S.J., A.C.E.), Montreal Neurological Institute, McGill University, Quebec, Canada
| | - Seok Jong Chung
- From the Departments of Neurology (S.W.K., H.S.Y., S.J.C., P.H.L., Y.H.S., B.S.Y.) and Nuclear Medicine (M.Y.), Yonsei University College of Medicine, Seoul, Korea; and McGill Centre for Integrative Neuroscience (S.J., A.C.E.), Montreal Neurological Institute, McGill University, Quebec, Canada
| | - Phil Hyu Lee
- From the Departments of Neurology (S.W.K., H.S.Y., S.J.C., P.H.L., Y.H.S., B.S.Y.) and Nuclear Medicine (M.Y.), Yonsei University College of Medicine, Seoul, Korea; and McGill Centre for Integrative Neuroscience (S.J., A.C.E.), Montreal Neurological Institute, McGill University, Quebec, Canada
| | - Young H Sohn
- From the Departments of Neurology (S.W.K., H.S.Y., S.J.C., P.H.L., Y.H.S., B.S.Y.) and Nuclear Medicine (M.Y.), Yonsei University College of Medicine, Seoul, Korea; and McGill Centre for Integrative Neuroscience (S.J., A.C.E.), Montreal Neurological Institute, McGill University, Quebec, Canada
| | - Mijin Yun
- From the Departments of Neurology (S.W.K., H.S.Y., S.J.C., P.H.L., Y.H.S., B.S.Y.) and Nuclear Medicine (M.Y.), Yonsei University College of Medicine, Seoul, Korea; and McGill Centre for Integrative Neuroscience (S.J., A.C.E.), Montreal Neurological Institute, McGill University, Quebec, Canada
| | - Alan C Evans
- From the Departments of Neurology (S.W.K., H.S.Y., S.J.C., P.H.L., Y.H.S., B.S.Y.) and Nuclear Medicine (M.Y.), Yonsei University College of Medicine, Seoul, Korea; and McGill Centre for Integrative Neuroscience (S.J., A.C.E.), Montreal Neurological Institute, McGill University, Quebec, Canada
| | - Byoung Seok Ye
- From the Departments of Neurology (S.W.K., H.S.Y., S.J.C., P.H.L., Y.H.S., B.S.Y.) and Nuclear Medicine (M.Y.), Yonsei University College of Medicine, Seoul, Korea; and McGill Centre for Integrative Neuroscience (S.J., A.C.E.), Montreal Neurological Institute, McGill University, Quebec, Canada.
| |
Collapse
|
49
|
Gratwicke JP, Foltynie T. Early nucleus basalis of Meynert degeneration predicts cognitive decline in Parkinson's disease. Brain 2019; 141:7-10. [PMID: 29325047 DOI: 10.1093/brain/awx333] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- James P Gratwicke
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK.,Department of Neurology, King's College Hospital, London, SE5 9RS, UK
| | - Thomas Foltynie
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| |
Collapse
|
50
|
Neuroprotective Actions of Glucagon-Like Peptide-1 (GLP-1) Analogues in Alzheimer's and Parkinson's Diseases. CNS Drugs 2019; 33:209-223. [PMID: 30511349 DOI: 10.1007/s40263-018-0593-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The current absence of effective treatments for Alzheimer's disease (AD) and Parkinson's disease (PD) reflects an incomplete knowledge of the underlying disease processes. Considerable efforts have been made to investigate the central pathological features of these diseases, giving rise to numerous attempts to develop compounds that interfere with such features. However, further characterization of the molecular targets within the interconnected AD and PD pathways is still required. Impaired brain insulin signaling has emerged as a feature that contributes to neuronal dysfunction in both AD and PD, leading to strategies aiming at restoring this pathway in the brain. Long-acting glucagon-like peptide-1 (GLP-1) analogues marketed for treatment of type 2 diabetes mellitus have been tested and have shown encouraging protective actions in experimental models of AD and PD as well as in initial clinical trials. We review studies revealing the neuroprotective actions of GLP-1 analogues in pre-clinical models of AD and PD and promising results from recent clinical trials.
Collapse
|